1
|
Li X, Li Y, Xiong B, Qiu S. Progress of Antimicrobial Mechanisms of Stilbenoids. Pharmaceutics 2024; 16:663. [PMID: 38794325 PMCID: PMC11124934 DOI: 10.3390/pharmaceutics16050663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
Antimicrobial drugs have made outstanding contributions to the treatment of pathogenic infections. However, the emergence of drug resistance continues to be a major threat to human health in recent years, and therefore, the search for novel antimicrobial drugs is particularly urgent. With a deeper understanding of microbial habits and drug resistance mechanisms, various creative strategies for the development of novel antibiotics have been proposed. Stilbenoids, characterized by a C6-C2-C6 carbon skeleton, have recently been widely recognized for their flexible antimicrobial roles. Here, we comprehensively summarize the mode of action of stilbenoids from the viewpoint of their direct antimicrobial properties, antibiofilm and antivirulence activities and their role in reversing drug resistance. This review will provide an important reference for the future development and research into the mechanisms of stilbenoids as antimicrobial agents.
Collapse
Affiliation(s)
- Xiancai Li
- Key Laboratory of Plant Resources Conservation and Sustainable Utilization, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou 510650, China;
| | - Yongqing Li
- Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou 510650, China;
| | - Binghong Xiong
- Key Laboratory of Plant Resources Conservation and Sustainable Utilization, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou 510650, China;
| | - Shengxiang Qiu
- Key Laboratory of Plant Resources Conservation and Sustainable Utilization, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou 510650, China;
| |
Collapse
|
2
|
Schmidt K, Scholz HC, Appelt S, Michel J, Jacob D, Dupke S. Virulence and resistance patterns of Vibrio cholerae non-O1/non-O139 acquired in Germany and other European countries. Front Microbiol 2023; 14:1282135. [PMID: 38075873 PMCID: PMC10703170 DOI: 10.3389/fmicb.2023.1282135] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 10/30/2023] [Indexed: 01/25/2025] Open
Abstract
Global warming has caused an increase in the emergence of Vibrio species in marine and estuarine environments as well as fresh water bodies. Over the past decades, antimicrobial resistance (AMR) has evolved among Vibrio species toward various antibiotics commonly used for the treatment of Vibrio infections. In this study, we assessed virulence and resistance patterns of Vibrio cholerae non-O1/non-O139 strains derived from Germany and other European countries. A total of 63 clinical and 24 environmental Vibrio cholerae non-O1/non-O139 strains, collected between 2011 and 2021, were analyzed. In silico antibiotic resistances were compared with resistance phenotypes according to EUCAST breakpoints. Additionally, genetic relatedness between isolates was assessed by two cgMLST schemes (SeqSphere +, pubMLST). Both cgMLST schemes yielded similar results, indicating high genetic diversity among V. cholerae non-O1/non-O139 isolates. Some isolates were found to be genetically closely related (allelic distance < 20), which suggests an epidemiological link. Thirty-seven virulence genes (VGs) were identified among 87 V. cholerae non-O1/non-O139 isolates, which resulted in 38 virulence profiles (VPs). VPs were similar between clinical and environmental isolates, with the exception of one clinical isolate that displayed a higher abundance of VGs. Also, a cluster of 11 environmental isolates was identified to have the lowest number of VGs. Among all strains, the predominant virulence factors were quorum sensing protein (luxS), repeats-in-toxins (rtxC/rtxD), hemolysin (hlyA) and different type VI secretion systems (T6SS) genes. The genotypic profiles revealed antibiotic resistance genes (ARGs) associated with resistance to beta-lactams, quinolones, macrolides, tetracycline, antifolate, aminoglycosides, fosfomycin, phenicols and sulfonamide. Carbapenemase gene VCC-1 was detected in 10 meropenem-resistant V. cholerae non-O1/non-O139 isolates derived from surface water in Germany. The proportion of resistance among V. cholerae non-O1/non-O139 species isolates against first line treatment (3rd generation cephalosporin, tetracycline and fluoroquinolone) was low. Empirical treatment would likely have been effective for all of the clinical V. cholerae non-O1/non-O139 isolates examined. Nevertheless, carbapenem-resistant isolates have been present in fresh water in Germany and might represent a reservoir for ARGs. Monitoring antimicrobial resistance is crucial for public health authorities to minimize the risks for the human population.
Collapse
Affiliation(s)
- Katarzyna Schmidt
- Mycotic and Parasitic Agents and Mycobacteria, Robert Koch Institute, Berlin, Germany
- ECDC Fellowship Programme, Public Health Microbiology Path (EUPHEM), European Centre for Disease Prevention and Control (ECDC), Stockholm, Sweden
| | - Holger C Scholz
- Centre for Biological Threats and Special Pathogens, Highly Pathogenic Microorganisms (ZBS 2), Robert Koch Institute, Berlin, Germany
| | - Sandra Appelt
- Centre for Biological Threats and Special Pathogens, Highly Pathogenic Microorganisms (ZBS 2), Robert Koch Institute, Berlin, Germany
| | - Jana Michel
- Centre for Biological Threats and Special Pathogens, Highly Pathogenic Microorganisms (ZBS 2), Robert Koch Institute, Berlin, Germany
| | - Daniela Jacob
- Centre for Biological Threats and Special Pathogens, Highly Pathogenic Microorganisms (ZBS 2), Robert Koch Institute, Berlin, Germany
| | - Susann Dupke
- Centre for Biological Threats and Special Pathogens, Highly Pathogenic Microorganisms (ZBS 2), Robert Koch Institute, Berlin, Germany
| |
Collapse
|
3
|
Nuttall RA, Moisander PH. Vibrio cyclitrophicus population-specific biofilm formation and epibiotic growth on marine copepods. Environ Microbiol 2023; 25:2534-2548. [PMID: 37612139 DOI: 10.1111/1462-2920.16483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 07/31/2023] [Indexed: 08/25/2023]
Abstract
Vibrio spp. form a part of the microbiome of copepods-an abundant component of marine mesozooplankton. The biological mechanisms of the Vibrio-copepod association are largely unknown. In this study we compared biofilm formation of V. cyclitrophicus isolated from copepods (L-strains related to other particle-associated strains) and closely related strains originating from seawater (S-strains), and visualized and quantified their attachment and growth on copepods. The S- and L-strains formed similar biofilms in the presence of complete sea salts, suggesting previously unknown biofilm mechanisms in the S-strains. No biofilms formed if sodium chloride was present as the only salt but added calcium significantly enhanced biofilms in the L-strains. GFP-L-strain cells attached to live copepods at higher numbers than the S-strains, suggesting distinct mechanisms, potentially including calcium, support their colonization of copepods. The cells grew on live copepods after attachment, demonstrating that copepods sustain epibiotic V. cyclitrophicus growth in situ. The results demonstrate that in spite of their 99.1% average nucleotide identity, these V. cyclitrophicus strains have a differential capacity to colonize marine copepods. The introduced V. cyclitrophicus-A. tonsa model could be informative in future studies on Vibrio-copepod association.
Collapse
Affiliation(s)
- Ryan A Nuttall
- Department of Biology, University of Massachusetts Dartmouth, North Dartmouth, Massachusetts, USA
| | - Pia H Moisander
- Department of Biology, University of Massachusetts Dartmouth, North Dartmouth, Massachusetts, USA
| |
Collapse
|
4
|
Lloyd CJ, Guo S, Kinrade B, Zahiri H, Eves R, Ali SK, Yildiz F, Voets IK, Davies PL, Klose KE. A peptide-binding domain shared with an Antarctic bacterium facilitates Vibrio cholerae human cell binding and intestinal colonization. Proc Natl Acad Sci U S A 2023; 120:e2308238120. [PMID: 37729203 PMCID: PMC10523503 DOI: 10.1073/pnas.2308238120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 07/31/2023] [Indexed: 09/22/2023] Open
Abstract
Vibrio cholerae, the causative agent of the disease cholera, is responsible for multiple pandemics. V. cholerae binds to and colonizes the gastrointestinal tract within the human host, as well as various surfaces in the marine environment (e.g., zooplankton) during interepidemic periods. A large adhesin, the Flagellar Regulated Hemagglutinin A (FrhA), enhances binding to erythrocytes and epithelial cells and enhances intestinal colonization. We identified a peptide-binding domain (PBD) within FrhA that mediates hemagglutination, binding to epithelial cells, intestinal colonization, and facilitates biofilm formation. Intriguingly, this domain is also found in the ice-binding protein of the Antarctic bacterium Marinomonas primoryensis, where it mediates binding to diatoms. Peptide inhibitors of the M. primoryensis PBD inhibit V. cholerae binding to human cells as well as to diatoms and inhibit biofilm formation. Moreover, the M. primoryensis PBD inserted into FrhA allows V. cholerae to bind human cells and colonize the intestine and also enhances biofilm formation, demonstrating the interchangeability of the PBD from these bacteria. Importantly, peptide inhibitors of PBD reduce V. cholerae intestinal colonization in infant mice. These studies demonstrate how V. cholerae uses a PBD shared with a diatom-binding Antarctic bacterium to facilitate intestinal colonization in humans and biofilm formation in the environment.
Collapse
Affiliation(s)
- Cameron J. Lloyd
- South Texas Center for Emerging Infectious Diseases, University of Texas, San Antonio, TX78249
- Department of Molecular Microbiology and Immunology, University of Texas, San Antonio, TX78249
| | - Shuaiqi Guo
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ONK7L 3N6, Canada
| | - Brett Kinrade
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ONK7L 3N6, Canada
| | - Hossein Zahiri
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ONK7L 3N6, Canada
| | - Robert Eves
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ONK7L 3N6, Canada
| | - Syed Khalid Ali
- South Texas Center for Emerging Infectious Diseases, University of Texas, San Antonio, TX78249
- Department of Molecular Microbiology and Immunology, University of Texas, San Antonio, TX78249
| | - Fitnat Yildiz
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, CA95064
| | - Ilja K. Voets
- Laboratory of Macromolecular and Organic Chemistry, Eindhoven University of Technology, Eindhoven5612, the Netherlands
| | - Peter L. Davies
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ONK7L 3N6, Canada
| | - Karl E. Klose
- South Texas Center for Emerging Infectious Diseases, University of Texas, San Antonio, TX78249
- Department of Molecular Microbiology and Immunology, University of Texas, San Antonio, TX78249
| |
Collapse
|
5
|
Zhao D, Ali A, Zuck C, Uy L, Morris JG, Wong ACN. Vibrio cholerae Invasion Dynamics of the Chironomid Host Are Strongly Influenced by Aquatic Cell Density and Can Vary by Strain. Microbiol Spectr 2023; 11:e0265222. [PMID: 37074192 PMCID: PMC10269514 DOI: 10.1128/spectrum.02652-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 03/24/2023] [Indexed: 04/20/2023] Open
Abstract
Cholera has been a human scourge since the early 1800s and remains a global public health challenge, caused by the toxigenic strains of the bacterium Vibrio cholerae. In its aquatic reservoirs, V. cholerae has been shown to live in association with various arthropod hosts, including the chironomids, a diverse insect family commonly found in wet and semiwet habitats. The association between V. cholerae and chironomids may shield the bacterium from environmental stressors and amplify its dissemination. However, the interaction dynamics between V. cholerae and chironomids remain largely unknown. In this study, we developed freshwater microcosms with chironomid larvae to test the effects of cell density and strain on V. cholerae-chironomid interactions. Our results show that chironomid larvae can be exposed to V. cholerae up to a high inoculation dose (109 cells/mL) without observable detrimental effects. Meanwhile, interstrain variability in host invasion, including prevalence, bacterial load, and effects on host survival, was highly cell density-dependent. Microbiome analysis of the chironomid samples by 16S rRNA gene amplicon sequencing revealed a general effect of V. cholerae exposure on microbiome species evenness. Taken together, our results provide novel insights into V. cholerae invasion dynamics of the chironomid larvae with respect to various doses and strains. The findings suggest that aquatic cell density is a crucial driver of V. cholerae invasion success in chironomid larvae and pave the way for future work examining the effects of a broader dose range and environmental variables (e.g., temperature) on V. cholerae-chironomid interactions. IMPORTANCE Vibrio cholerae is the causative agent of cholera, a significant diarrheal disease affecting millions of people worldwide. Increasing evidence suggests that the environmental facets of the V. cholerae life cycle involve symbiotic associations with aquatic arthropods, which may facilitate its environmental persistence and dissemination. However, the dynamics of interactions between V. cholerae and aquatic arthropods remain unexplored. This study capitalized on using freshwater microcosms with chironomid larvae to investigate the effects of bacterial cell density and strain on V. cholerae-chironomid interactions. Our results suggest that aquatic cell density is the primary determinant of V. cholerae invasion success in chironomid larvae, while interstrain variability in invasion outcomes can be observed under specific cell density conditions. We also determined that V. cholerae exposure generally reduces species evenness of the chironomid-associated microbiome. Collectively, these findings provide novel insights into V. cholerae-arthropod interactions using a newly developed experimental host system.
Collapse
Affiliation(s)
- Dianshu Zhao
- Entomology and Nematology Department, University of Florida, Gainesville, Florida, USA
| | - Afsar Ali
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, Florida, USA
| | - Cameron Zuck
- Entomology and Nematology Department, University of Florida, Gainesville, Florida, USA
| | - Laurice Uy
- Entomology and Nematology Department, University of Florida, Gainesville, Florida, USA
| | - J. Glenn Morris
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
| | - Adam Chun-Nin Wong
- Entomology and Nematology Department, University of Florida, Gainesville, Florida, USA
- Genetics Institute, University of Florida, Gainesville, Florida, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
6
|
Vidakovic L, Mikhaleva S, Jeckel H, Nisnevich V, Strenger K, Neuhaus K, Raveendran K, Ben-Moshe NB, Aznaourova M, Nosho K, Drescher A, Schmeck B, Schulte LN, Persat A, Avraham R, Drescher K. Biofilm formation on human immune cells is a multicellular predation strategy of Vibrio cholerae. Cell 2023; 186:2690-2704.e20. [PMID: 37295405 PMCID: PMC10256282 DOI: 10.1016/j.cell.2023.05.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 01/26/2023] [Accepted: 05/09/2023] [Indexed: 06/12/2023]
Abstract
Biofilm formation is generally recognized as a bacterial defense mechanism against environmental threats, including antibiotics, bacteriophages, and leukocytes of the human immune system. Here, we show that for the human pathogen Vibrio cholerae, biofilm formation is not only a protective trait but also an aggressive trait to collectively predate different immune cells. We find that V. cholerae forms biofilms on the eukaryotic cell surface using an extracellular matrix comprising primarily mannose-sensitive hemagglutinin pili, toxin-coregulated pili, and the secreted colonization factor TcpF, which differs from the matrix composition of biofilms on other surfaces. These biofilms encase immune cells and establish a high local concentration of a secreted hemolysin to kill the immune cells before the biofilms disperse in a c-di-GMP-dependent manner. Together, these results uncover how bacteria employ biofilm formation as a multicellular strategy to invert the typical relationship between human immune cells as the hunters and bacteria as the hunted.
Collapse
Affiliation(s)
| | - Sofya Mikhaleva
- Institute of Bioengineering and Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Hannah Jeckel
- Biozentrum, University of Basel, 4056 Basel, Switzerland; Department of Physics, Philipps-Universität Marburg, 35043 Marburg, Germany
| | - Valerya Nisnevich
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | | | - Konstantin Neuhaus
- Biozentrum, University of Basel, 4056 Basel, Switzerland; Department of Physics, Philipps-Universität Marburg, 35043 Marburg, Germany
| | | | - Noa Bossel Ben-Moshe
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Marina Aznaourova
- Institute for Lung Research, Center for Synthetic Microbiology (SYNMIKRO), Universities of Giessen and Marburg Lung Center, Philipps-Universität Marburg, 35043 Marburg, Germany
| | - Kazuki Nosho
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Antje Drescher
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Bernd Schmeck
- Institute for Lung Research, Center for Synthetic Microbiology (SYNMIKRO), Universities of Giessen and Marburg Lung Center, Philipps-Universität Marburg, 35043 Marburg, Germany; Department of Pulmonary and Critical Care Medicine, University Medical Center Marburg, 35043 Marburg, Germany; German Center for Infection Research (DZIF), 35043 Marburg, Germany; German Center for Lung Research (DZL), 35043 Marburg, Germany; Institute for Lung Health, 35392 Giessen, Germany
| | - Leon N Schulte
- Institute for Lung Research, Center for Synthetic Microbiology (SYNMIKRO), Universities of Giessen and Marburg Lung Center, Philipps-Universität Marburg, 35043 Marburg, Germany; German Center for Lung Research (DZL), 35043 Marburg, Germany
| | - Alexandre Persat
- Institute of Bioengineering and Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Roi Avraham
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Knut Drescher
- Biozentrum, University of Basel, 4056 Basel, Switzerland.
| |
Collapse
|
7
|
Sarveswari HB, Gupta KK, Durai R, Solomon AP. Development of a smart pH-responsive nano-polymer drug, 2-methoxy-4-vinylphenol conjugate against the intestinal pathogen, Vibrio cholerae. Sci Rep 2023; 13:1250. [PMID: 36690664 PMCID: PMC9871008 DOI: 10.1038/s41598-023-28033-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 01/11/2023] [Indexed: 01/24/2023] Open
Abstract
Vibrio cholerae causes cholera, an acute diarrhoeal disease. The virulence in V. cholerae is regulated by the quorum-sensing mechanism and response regulator LuxO positively regulates the expression of virulence determinants adhesion, biofilm formation, and cholera toxin production. Previous in-silico studies revealed that 2-methoxy-4-vinylphenol could bind to the ATP binding site of LuxO and the complex was compact and stable in pHs like intestinal pHs. Here, we have explored the polymeric nano-formulation of 2-methoxy-4-vinylphenol using cellulose acetate phthalate for controlled drug release and their effectiveness in attenuating the expression of V. cholerae virulence. Physico-chemical characterization of the formulation showed particles with a mean size of 91.8 ± 14 nm diameter and surface charge of - 14.7 ± 0.07 mV. The uniform round polymeric nanoparticles formed displayed about 51% burst release of the drug at pH 7 by 3rd h, followed by a controlled linear release in alkaline pH. The polymeric nanoparticles demonstrated a tenfold increase in intestinal membrane permeability ex-vivo. At lower concentrations, the 2-methoxy-4-vinylphenol polymeric nanoparticles were non-cytotoxic to Int 407 cells. In-vitro analysis at pH 6, pH 7, pH 8, and pH 9 revealed that cellulose acetate phthalate-2-methoxy-4-vinylphenol nanoparticles were non-bactericidal at concentrations up to 500 μg/mL. At 31.25 μg/mL, the nanoparticles inhibited about 50% of the biofilm formation of V. cholerae MTCC 3905 and HYR14 strains. At this concentration, the adherence of V. cholerae MTCC 3905 and HYR14 to Int 407 cell lines were also significantly affected. Gene expression analysis revealed that the expression of tcp, qrr, and ct at pH 6, 7, 8, and 9 has reduced. The CAP-2M4VP nanoparticles have demonstrated the potential to effectively reduce the virulence of V. cholerae in-vitro.
Collapse
Affiliation(s)
- Hema Bhagavathi Sarveswari
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, 613401, India
| | - Krishna Kant Gupta
- School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, 613401, India
| | - Ramyadevi Durai
- Pharmaceutical Technology Laboratory, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, 613401, India.
| | - Adline Princy Solomon
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, 613401, India.
| |
Collapse
|
8
|
Kim EJ, Bae J, Ju YJ, Ju DB, Lee D, Son S, Choi H, Ramamurthy T, Yun CH, Kim DW. Inactivated Vibrio cholerae Strains That Express TcpA via the toxT-139F Allele Induce Antibody Responses against TcpA. J Microbiol Biotechnol 2022; 32:1396-1405. [PMID: 36317425 PMCID: PMC9720071 DOI: 10.4014/jmb.2209.09001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/04/2022] [Accepted: 10/06/2022] [Indexed: 11/06/2022]
Abstract
Cholera remains a major global public health problem, for which oral cholera vaccines (OCVs) being a valuable strategy. Patients, who have recovered from cholera, develop antibody responses against LPS, cholera toxin (CT), toxin-coregulated pilus (TCP) major subunit A (TcpA) and other antigens; thus, these responses are potentially important contributors to immunity against Vibrio cholerae infection. However, assessments of the efficacy of current OCVs, especially inactivated OCVs, have focused primarily on O-antigen-specific antibody responses, suggesting that more sophisticated strategies are required for inactivated OCVs to induce immune responses against TCP, CT, and other antigens. Previously, we have shown that the toxT-139F allele enables V. cholerae strains to produce CT and TCP under simple laboratory culture conditions. Thus, we hypothesized that V. cholerae strains that express TCP via the toxT-139F allele induce TCP-specific antibody responses. As anticipated, V. cholerae strains that expressed TCP through the toxT-139F allele elicited antibody responses against TCP when the inactivated bacteria were delivered via a mouse model. We have further developed TCP-expressing V. cholerae strains that have been used in inactivated OCVs and shown that they effect an antibody response against TcpA in vivo, suggesting that V. cholerae strains with the toxT-139F allele are excellent candidates for cholera vaccines.
Collapse
Affiliation(s)
- Eun Jin Kim
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan 15588, Republic of Korea,Institute of Pharmacological Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Jonghyun Bae
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan 15588, Republic of Korea,Institute of Pharmacological Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Young-Jun Ju
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Do-Bin Ju
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Donghyun Lee
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan 15588, Republic of Korea,Institute of Pharmacological Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Seonghyeon Son
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan 15588, Republic of Korea,Institute of Pharmacological Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Hunseok Choi
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan 15588, Republic of Korea,Institute of Pharmacological Research, Hanyang University, Ansan 15588, Republic of Korea
| | | | - Cheol-Heui Yun
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Republic of Korea,Corresponding authors C.-H. Yun Phone: + 82-2-880-4802 E-mail:
| | - Dong Wook Kim
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan 15588, Republic of Korea,Institute of Pharmacological Research, Hanyang University, Ansan 15588, Republic of Korea,
D.W. Kim Phone: +82-31-400-5806 E-mail:
| |
Collapse
|
9
|
Recombinant Globular Domain of TcpA Pilin from Vibrio cholerae El Tor: Recovery from Inclusion Bodies and Structural Characterization. Life (Basel) 2022; 12:life12111802. [DOI: 10.3390/life12111802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/28/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
The production of recombinant proteins in Escherichia coli cells is often hampered by aggregation of newly synthesized proteins and formation of inclusion bodies. Here we propose the use of transverse urea gradient electrophoresis (TUGE) in testing the capability of folding of a recombinant protein from inclusion bodies dissolved in urea. A plasmid encoding the amino acid sequence 55–224 of TcpA pilin (C-terminal globular domain: TcpA-C) from Vibrio cholerae El Tor enlarged by a His-tag on its N-terminus was expressed in E. coli cells. The major fraction (about 90%) of the target polypeptide was detected in cell debris. The polypeptide was isolated from the soluble fraction and recovered from inclusion bodies after their urea treatment. Some structural properties of the polypeptide from each sample proved identical. The refolding protocol was developed on the basis of TUGE data and successfully used for the protein large-scale recovery from inclusion bodies. Spectral, hydrodynamic, and thermodynamic characteristics of the recombinant TcpA recovered from inclusion bodies indicate the presence of a globular conformation with a pronounced secondary structure and a rigid tertiary structure, which is promising for the design of immunodiagnostics preparations aimed to assess the pilin level in different strains of V. cholerae and to develop cholera vaccines.
Collapse
|
10
|
Oki H, Kawahara K, Iimori M, Imoto Y, Nishiumi H, Maruno T, Uchiyama S, Muroga Y, Yoshida A, Yoshida T, Ohkubo T, Matsuda S, Iida T, Nakamura S. Structural basis for the toxin-coregulated pilus-dependent secretion of Vibrio cholerae colonization factor. SCIENCE ADVANCES 2022; 8:eabo3013. [PMID: 36240278 PMCID: PMC9565799 DOI: 10.1126/sciadv.abo3013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 08/26/2022] [Indexed: 06/16/2023]
Abstract
Colonization of the host intestine is the most important step in Vibrio cholerae infection. The toxin-coregulated pilus (TCP), an operon-encoded type IVb pilus (T4bP), plays a crucial role in this process, which requires an additional secreted protein, TcpF, encoded on the same TCP operon; however, its mechanisms of secretion and function remain elusive. Here, we demonstrated that TcpF interacts with the minor pilin, TcpB, of TCP and elucidated the crystal structures of TcpB alone and in complex with TcpF. The structural analyses reveal how TCP recognizes TcpF and its secretory mechanism via TcpB-dependent pilus elongation and retraction. Upon binding to TCP, TcpF forms a flower-shaped homotrimer with its flexible N terminus hooked onto the trimeric interface of TcpB. Thus, the interaction between the minor pilin and the N terminus of the secreted protein, namely, the T4bP secretion signal, is key for V. cholerae colonization and is a new potential therapeutic target.
Collapse
Affiliation(s)
- Hiroya Oki
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Kazuki Kawahara
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
| | - Minato Iimori
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Yuka Imoto
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Haruka Nishiumi
- Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Takahiro Maruno
- Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Susumu Uchiyama
- Graduate School of Engineering, Osaka University, Osaka, Japan
- Department of Creative Research, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Aichi, Japan
- U-Medico Inc., Suita, Osaka, Japan
| | - Yuki Muroga
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Akihiro Yoshida
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Takuya Yoshida
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Tadayasu Ohkubo
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
| | - Shigeaki Matsuda
- Department of Bacterial Infections, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Tetsuya Iida
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
- Department of Bacterial Infections, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Shota Nakamura
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
| |
Collapse
|
11
|
Khouadja S, Roque A, Gonzalez M, Furones D. Vibrio pathogenicity island and phage CTX genes in Vibrio alginolyticus isolated from different aquatic environments. JOURNAL OF WATER AND HEALTH 2022; 20:1469-1478. [PMID: 36308492 DOI: 10.2166/wh.2022.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
In the present study, we investigated the presence of four Vibrio cholerae virulence genes (ctxA, VPI, Zot and ace) in 36 Vibrio alginolyticus isolates obtained from different seawater, sediments and aquatic organisms. We tested the virulence of 13 V. alginolyticus strains against juveniles of Sparus aurata and this virulence was correlated with the presence of V. cholerae virulence genes. A positive amplification for the virulence pathogenicity island was produced by five V. alginolyticus strains and four for cholerae toxin. Some of the V. alginolyticus strains are pathogenic to aquatic animals and might have derived their virulence genes from V. cholerae. V. alginolyticus strains can be considered as a possible reservoir of V. cholerae virulence genes.
Collapse
Affiliation(s)
- Sadok Khouadja
- Laboratoire d'Analyse, Traitement et Valorisation des Polluants de l'Environnement et des Produits, Département de Microbiologie, Faculté de Pharmacie, Rue Avicenne 5000, Monastir, Tunisia E-mail:
| | - Ana Roque
- IRTA-SCR, Ctra. Poble Nou Km 7.5, 43540 Sant Carles de la Ràpita, Tarragona, Spain
| | - Mar Gonzalez
- IRTA-SCR, Ctra. Poble Nou Km 7.5, 43540 Sant Carles de la Ràpita, Tarragona, Spain
| | - Dolors Furones
- IRTA-SCR, Ctra. Poble Nou Km 7.5, 43540 Sant Carles de la Ràpita, Tarragona, Spain
| |
Collapse
|
12
|
Abstract
Cholera is a severe diarrheal disease caused by the bacterium Vibrio cholerae and constitutes a significant public health threat in many areas of the world. V. cholerae infection elicits potent and long-lasting immunity, and efforts to develop cholera vaccines have been ongoing for more than a century. Currently available inactivated two-dose oral cholera vaccines are increasingly deployed to both prevent and actively curb cholera outbreaks, and they are key components of the global effort to eradicate cholera. However, these killed whole-cell vaccines have several limitations, and a variety of new oral and nonoral cholera vaccine platforms have recently been developed. Here, we review emerging concepts in cholera vaccine design and implementation that have been driven by insights from human and animal studies. As a prototypical vaccine-preventable disease, cholera continues to be an excellent target for the development and application of cutting-edge technologies and platforms that may transform vaccinology. Expected final online publication date for the Annual Review of Microbiology, Volume 76 is September 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Brandon Sit
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, Massachusetts, USA; .,Department of Microbiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Bolutife Fakoya
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, Massachusetts, USA; .,Department of Microbiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Matthew K Waldor
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, Massachusetts, USA; .,Department of Microbiology, Harvard Medical School, Boston, Massachusetts, USA.,Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Massachusetts, USA.,Howard Hughes Medical Institute, Bethesda, Maryland, USA
| |
Collapse
|
13
|
Wang G, Mohanty B, Williams ML, Doak BC, Dhouib R, Totsika M, McMahon R, Sharma G, Zheng D, Bentley MR, Chin YKY, Horne J, Chalmers DK, Heras B, Scanlon MJ. Selective binding of small molecules to Vibrio cholerae DsbA offers a starting point for the design of novel antibacterials. ChemMedChem 2022; 17:e202100673. [PMID: 34978144 PMCID: PMC9305425 DOI: 10.1002/cmdc.202100673] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/23/2021] [Indexed: 11/25/2022]
Abstract
DsbA enzymes catalyze oxidative folding of proteins that are secreted into the periplasm of Gram‐negative bacteria, and they are indispensable for the virulence of human pathogens such as Vibrio cholerae and Escherichia coli. Therefore, targeting DsbA represents an attractive approach to control bacterial virulence. X‐ray crystal structures reveal that DsbA enzymes share a similar fold, however, the hydrophobic groove adjacent to the active site, which is implicated in substrate binding, is shorter and flatter in the structure of V. cholerae DsbA (VcDsbA) compared to E. coli DsbA (EcDsbA). The flat and largely featureless nature of this hydrophobic groove is challenging for the development of small molecule inhibitors. Using fragment‐based screening approaches, we have identified a novel small molecule, based on the benzimidazole scaffold, that binds to the hydrophobic groove of oxidized VcDsbA with a KD of 446±10 μM. The same benzimidazole compound has ∼8‐fold selectivity for VcDsbA over EcDsbA and binds to oxidized EcDsbA, with KD>3.5 mM. We generated a model of the benzimidazole complex with VcDsbA using NMR data but were unable to determine the structure of the benzimidazole bound EcDsbA using either NMR or X‐ray crystallography. Therefore, a structural basis for the observed selectivity is unclear. To better understand ligand binding to these two enzymes we crystallized each of them in complex with a known ligand, the bile salt sodium taurocholate. The crystal structures show that taurocholate adopts different binding poses in complex with VcDsbA and EcDsbA, and reveal the protein‐ligand interactions that stabilize the different modes of binding. This work highlights the capacity of fragment‐based drug discovery to identify inhibitors of challenging protein targets. In addition, it provides a starting point for development of more potent and specific VcDsbA inhibitors that act through a novel anti‐virulence mechanism.
Collapse
Affiliation(s)
- Geqing Wang
- La Trobe University - Bundoora Campus: La Trobe University, Department of Biochemistry and Genetics, AUSTRALIA
| | | | - Martin L Williams
- Monash Institute of Pharmaceutical Sciences, Medicinal Chemistry, AUSTRALIA
| | - Bradley C Doak
- Monash Institute of Pharmaceutical Sciences, Medicinal Chemistry, AUSTRALIA
| | - Rabeb Dhouib
- Queensland University of Technology, School of Biomedical Sciences, AUSTRALIA
| | - Makrina Totsika
- Queensland University of Technology, School of Biomedical Sciences, AUSTRALIA
| | - Roisin McMahon
- Griffith University, Griffith Institute for Drug Discovery, AUSTRALIA
| | - Gaurav Sharma
- Monash Institute of Pharmaceutical Sciences, Medicinal Chemistry, AUSTRALIA
| | - Dan Zheng
- Monash Institute of Pharmaceutical Sciences, Medicinal Chemistry, AUSTRALIA
| | - Matthew R Bentley
- Monash Institute of Pharmaceutical Sciences, Medicinal Chemistry, AUSTRALIA
| | - Yanni Ka-Yan Chin
- The University of Queensland, Cantre for Advanced Imaging, AUSTRALIA
| | - James Horne
- University of Tasmania, Central Science Laboratory, AUSTRALIA
| | - David K Chalmers
- Monash Institute of Pharmaceutical Sciences, Medicinal Chemistry, AUSTRALIA
| | - Begoña Heras
- La Trobe University, Department of Biochemistry and Genetics, AUSTRALIA
| | - Martin Joseph Scanlon
- Monash Institute of Pharmaceutical Sciences Monash University Parkville Campus, Medicinal Chemistry, 381 Royal Parade, Monash University, 3052, Parkville, AUSTRALIA
| |
Collapse
|
14
|
Nishiyama K, Yokoi T, Sugiyama M, Osawa R, Mukai T, Okada N. Roles of the Cell Surface Architecture of Bacteroides and Bifidobacterium in the Gut Colonization. Front Microbiol 2021; 12:754819. [PMID: 34721360 PMCID: PMC8551831 DOI: 10.3389/fmicb.2021.754819] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 09/24/2021] [Indexed: 12/12/2022] Open
Abstract
There are numerous bacteria reside within the mammalian gastrointestinal tract. Among the intestinal bacteria, Akkermansia, Bacteroides, Bifidobacterium, and Ruminococcus closely interact with the intestinal mucus layer and are, therefore, known as mucosal bacteria. Mucosal bacteria use host or dietary glycans for colonization via adhesion, allowing access to the carbon source that the host’s nutrients provide. Cell wall or membrane proteins, polysaccharides, and extracellular vesicles facilitate these mucosal bacteria-host interactions. Recent studies revealed that the physiological properties of Bacteroides and Bifidobacterium significantly change in the presence of co-existing symbiotic bacteria or markedly differ with the spatial distribution in the mucosal niche. These recently discovered strategic colonization processes are important for understanding the survival of bacteria in the gut. In this review, first, we introduce the experimental models used to study host-bacteria interactions, and then, we highlight the latest discoveries on the colonization properties of mucosal bacteria, focusing on the roles of the cell surface architecture regarding Bacteroides and Bifidobacterium.
Collapse
Affiliation(s)
- Keita Nishiyama
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Tatsunari Yokoi
- Department of Microbiology, School of Pharmacy, Kitasato University, Tokyo, Japan
| | - Makoto Sugiyama
- Laboratory of Veterinary Anatomy, School of Veterinary Medicine, Kitasato University, Towada, Japan
| | - Ro Osawa
- Research Center for Food Safety and Security, Kobe University, Kobe, Japan
| | - Takao Mukai
- Department of Animal Science, School of Veterinary Medicine, Kitasato University, Towada, Japan
| | - Nobuhiko Okada
- Department of Microbiology, School of Pharmacy, Kitasato University, Tokyo, Japan
| |
Collapse
|
15
|
Das S, Chourashi R, Mukherjee P, Gope A, Koley H, Dutta M, Mukhopadhyay AK, Okamoto K, Chatterjee NS. Multifunctional transcription factor CytR of Vibrio cholerae is important for pathogenesis. MICROBIOLOGY-SGM 2021; 166:1136-1148. [PMID: 33150864 DOI: 10.1099/mic.0.000949] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Vibrio cholerae, the Gram-negative facultative pathogen, resides in the aquatic environment and infects humans and causes diarrhoeagenic cholera. Although the environment differs drastically, V. cholerae thrives in both of these conditions aptly and chitinases play a vital role in their persistence and nutrient acquisition. Chitinases also play a role in V. cholerae pathogenesis. Chitinases and its downstream chitin utilization genes are regulated by sensor histidine kinase ChiS, which also plays a significant role in pathogenesis. Recent exploration suggests that CytR, a transcription factor of the LacI family in V. cholerae, also regulates chitinase secretion in environmental conditions. Since chitinases and chitinase regulator ChiS is involved in pathogenesis, CytR might also play a significant role in pathogenicity. However, the role of CytR in pathogenesis is yet to be known. This study explores the regulation of CytR on the activation of ChiS in the presence of mucin and its role in pathogenesis. Therefore, we created a CytR isogenic mutant strain of V. cholerae (CytR¯) and found considerably less β-hexosaminidase enzyme production, which is an indicator of ChiS activity. The CytR¯ strain greatly reduced the expression of chitinases chiA1 and chiA2 in mucin-supplemented media. Electron microscopy showed that the CytR¯ strain was aflagellate. The expression of flagellar-synthesis regulatory genes flrB, flrC and class III flagellar-synthesis genes were reduced in the CytR¯ strain. The isogenic CytR mutant showed less growth compared to the wild-type in mucin-supplemented media as well as demonstrated highly retarded motility and reduced mucin-layer penetration. The CytR mutant revealed decreased adherence to the HT-29 cell line. In animal models, reduced fluid accumulation and colonization were observed during infection with the CytR¯ strain due to reduced expression of ctxB, toxT and tcpA. Collectively these data suggest that CytR plays an important role in V. cholerae pathogenesis.
Collapse
Affiliation(s)
- Suman Das
- Division of Biochemistry, ICMR - National Institute of Cholera and Enteric Diseases, Kolkata-700010, India
| | - Rhishita Chourashi
- Division of Biochemistry, ICMR - National Institute of Cholera and Enteric Diseases, Kolkata-700010, India
| | - Priyadarshini Mukherjee
- Division of Bacteriology, ICMR - National Institute of Cholera and Enteric Diseases, Kolkata-700010, India
| | - Animesh Gope
- Division of Clinical Medicine, ICMR - National Institute of Cholera and Enteric Diseases, Kolkata-700010, India
| | - Hemanta Koley
- Division of Bacteriology, ICMR - National Institute of Cholera and Enteric Diseases, Kolkata-700010, India
| | - Moumita Dutta
- Division of Electron Microscopy, ICMR - National Institute of Cholera and Enteric Diseases, Kolkata-700010, India
| | - Asish K Mukhopadhyay
- Division of Bacteriology, ICMR - National Institute of Cholera and Enteric Diseases, Kolkata-700010, India
| | - Keinosuke Okamoto
- Collaborative Research Center of Okayama University for Infectious Diseases at NICED, Kolkata, India
| | - Nabendu Sekhar Chatterjee
- Division of Biochemistry, ICMR - National Institute of Cholera and Enteric Diseases, Kolkata-700010, India
| |
Collapse
|
16
|
Vibrio cholerae's mysterious Seventh Pandemic island (VSP-II) encodes novel Zur-regulated zinc starvation genes involved in chemotaxis and cell congregation. PLoS Genet 2021; 17:e1009624. [PMID: 34153031 PMCID: PMC8248653 DOI: 10.1371/journal.pgen.1009624] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 07/01/2021] [Accepted: 05/27/2021] [Indexed: 11/19/2022] Open
Abstract
Vibrio cholerae is the causative agent of cholera, a notorious diarrheal disease that is typically transmitted via contaminated drinking water. The current pandemic agent, the El Tor biotype, has undergone several genetic changes that include horizontal acquisition of two genomic islands (VSP-I and VSP-II). VSP presence strongly correlates with pandemicity; however, the contribution of these islands to V. cholerae's life cycle, particularly the 26-kb VSP-II, remains poorly understood. VSP-II-encoded genes are not expressed under standard laboratory conditions, suggesting that their induction requires an unknown signal from the host or environment. One signal that bacteria encounter under both host and environmental conditions is metal limitation. While studying V. cholerae's zinc-starvation response in vitro, we noticed that a mutant constitutively expressing zinc starvation genes (Δzur) congregates at the bottom of a culture tube when grown in a nutrient-poor medium. Using transposon mutagenesis, we found that flagellar motility, chemotaxis, and VSP-II encoded genes were required for congregation. The VSP-II genes encode an AraC-like transcriptional activator (VerA) and a methyl-accepting chemotaxis protein (AerB). Using RNA-seq and lacZ transcriptional reporters, we show that VerA is a novel Zur target and an activator of the nearby AerB chemoreceptor. AerB interfaces with the chemotaxis system to drive oxygen-dependent congregation and energy taxis. Importantly, this work suggests a functional link between VSP-II, zinc-starved environments, and energy taxis, yielding insights into the role of VSP-II in a metal-limited host or aquatic reservoir.
Collapse
|
17
|
Ghasemi M, Bakhshi B, Khashei R, Soudi S, Boustanshenas M. Vibrio cholerae toxin coregulated pilus provokes inflammatory responses in Coculture model of Caco-2 and peripheral blood mononuclear cells (PBMC) leading to increased colonization. Microbiol Immunol 2021; 65:238-244. [PMID: 33913531 DOI: 10.1111/1348-0421.12889] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 03/09/2021] [Accepted: 04/26/2021] [Indexed: 11/30/2022]
Abstract
The aim of this study was to assess the modulatory effect of TcpA in the expression of CEACAM1 adhesin molecule and IL-1, IL-8, and TNF-α pro-inflammatory cytokines in the Coculture model of Caco-2/PBMC (peripheral blood mononuclear cell) that can mimic the intestinal milieu. The TcpA gene from Vibrio cholerae ATCC14035 was cloned in pET-28a and transformed into Escherichia coli Bl-21. The recombinant TcpA-His6 protein was expressed and purified using Ni-column chromatography. The sequencing of transformed plasmid and Western blot analysis of purified protein confirmed the identity of rTcp. The cytotoxicity of different concentrations of recombinant protein for human colon carcinoma cell line (human colorectal adenocarcinoma cell [Caco-2 cell]) was assessed by MTT assay and showed viability of 92%, 82%, and 70%, for 10 µg/mL of TcpA after 24, 48, and 72 h, respectively. Co-cultures of Caco-2 and PBMCs were used to mimic the intestinal milieu and treated with different concentrations of rTcpA (1, 5, 10, and 50 µg/mL). Our data showed about 2.04-, 3.37-, 3.68-, and 42.7-fold increase in CEACAM1 gene expression, respectively, compared with the nontreated Caco-2/PBMC Coculture. Moreover, the expression of IL-1, IL-8, and TNF-α genes was significantly increased up to 15.75-, 7.04-, and 80.95-folds, respectively. In conclusion, V. cholerae TcpA induces statistically significant dose-dependent stimulatory effect on TNF-α, IL-,1, and IL-8 pro-inflammatory cytokines expression. Of these, TNF-α was much more affected which, consequently, elevated the CEACAM1 expression level in IECs. This suggests that TcpA protein is a critical effector as an inducer of increased adhesion potential of V. cholera as well as inflammatory responses of host intestinal tissue.
Collapse
Affiliation(s)
- Maryam Ghasemi
- Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Bita Bakhshi
- Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Reza Khashei
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sara Soudi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mina Boustanshenas
- Antimicrobial Resistance Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
18
|
Chac D, Dunmire CN, Singh J, Weil AA. Update on Environmental and Host Factors Impacting the Risk of Vibrio cholerae Infection. ACS Infect Dis 2021; 7:1010-1019. [PMID: 33844507 DOI: 10.1021/acsinfecdis.0c00914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Vibrio cholerae is the causative agent of cholera, a diarrheal disease that kills tens of thousands of people each year. Cholera is transmitted primarily by the ingestion of drinking water contaminated with fecal matter, and a safe water supply remains out of reach in many areas of the world. In this Review, we discuss host and environmental factors that impact the susceptibility to V. cholerae infection and the severity of disease.
Collapse
Affiliation(s)
- Denise Chac
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington 98109, United States
| | - Chelsea N. Dunmire
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington 98109, United States
| | - Jasneet Singh
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington 98109, United States
| | - Ana A. Weil
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington 98109, United States
| |
Collapse
|
19
|
Alkaline pH Increases Swimming Speed and Facilitates Mucus Penetration for Vibrio cholerae. J Bacteriol 2021; 203:JB.00607-20. [PMID: 33468594 PMCID: PMC8088521 DOI: 10.1128/jb.00607-20] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 01/05/2021] [Indexed: 01/05/2023] Open
Abstract
The diarrheal disease cholera is still a burden for populations in developing countries with poor sanitation. To develop effective vaccines and prevention strategies against Vibrio cholerae, we must understand the initial steps of infection leading to the colonization of the small intestine. Intestinal mucus is the first line of defense against intestinal pathogens. It acts as a physical barrier between epithelial tissues and the lumen that enteropathogens must overcome to establish a successful infection. We investigated the motile behavior of two Vibrio cholerae strains (El Tor C6706 and Classical O395) in mucus using single-cell tracking in unprocessed porcine intestinal mucus. We determined that V. cholerae can penetrate mucus using flagellar motility and that alkaline pH increases swimming speed and, consequently, improves mucus penetration. Microrheological measurements indicate that changes in pH between 6 and 8 (the physiological range for the human small intestine) had little effect on the viscoelastic properties of mucus. Finally, we determined that acidic pH promotes surface attachment by activating the mannose-sensitive hemagglutinin (MshA) pilus in V. cholerae El Tor C6706 without a measurable change in the total cellular concentration of the secondary messenger cyclic dimeric GMP (c-di-GMP). Overall, our results support the hypothesis that pH is an important factor affecting the motile behavior of V. cholerae and its ability to penetrate mucus. Therefore, changes in pH along the human small intestine may play a role in determining the preferred site for V. cholerae during infection. IMPORTANCE The diarrheal disease cholera is still a burden for populations in developing countries with poor sanitation. To develop effective vaccines and prevention strategies against Vibrio cholerae, we must understand the initial steps of infection leading to the colonization of the small intestine. To infect the host and deliver the cholera toxin, V. cholerae has to penetrate the mucus layer protecting the intestinal tissues. However, the interaction of V. cholerae with intestinal mucus has not been extensively investigated. In this report, we demonstrated using single-cell tracking that V. cholerae can penetrate intestinal mucus using flagellar motility. In addition, we observed that alkaline pH improves the ability of V. cholerae to penetrate mucus. This finding has important implications for understanding the dynamics of infection, because pH varies significantly along the small intestine, between individuals, and between species. Blocking mucus penetration by interfering with flagellar motility in V. cholerae, reinforcing the mucosa, controlling intestinal pH, or manipulating the intestinal microbiome will offer new strategies to fight cholera.
Collapse
|
20
|
Cho JY, Liu R, Macbeth JC, Hsiao A. The Interface of Vibrio cholerae and the Gut Microbiome. Gut Microbes 2021; 13:1937015. [PMID: 34180341 PMCID: PMC8244777 DOI: 10.1080/19490976.2021.1937015] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 05/18/2021] [Accepted: 05/24/2021] [Indexed: 02/04/2023] Open
Abstract
The bacterium Vibrio cholerae is the etiologic agent of the severe human diarrheal disease cholera. The gut microbiome, or the native community of microorganisms found in the human gastrointestinal tract, is increasingly being recognized as a factor in driving susceptibility to infection, in vivo fitness, and host interactions of this pathogen. Here, we review a subset of the emerging studies in how gut microbiome structure and microbial function are able to drive V. cholerae virulence gene regulation, metabolism, and modulate host immune responses to cholera infection and vaccination. Improved mechanistic understanding of commensal-pathogen interactions offers new perspectives in the design of prophylactic and therapeutic approaches for cholera control.
Collapse
Affiliation(s)
- Jennifer Y. Cho
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA, USA
- Department of Biochemistry, University of California, Riverside, California, USA
| | - Rui Liu
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA, USA
- Graduate Program in Genetics, Genomics, and Bioinformatics, University of California, Riverside, California, USA
| | - John C. Macbeth
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA, USA
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California, USA
| | - Ansel Hsiao
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA, USA
| |
Collapse
|
21
|
Ghasemi M, Bakhshi B, Khashei R, Soudi S. Modulatory effect of Vibrio cholerae toxin co-regulated pilus on mucins, toll-like receptors and NOD genes expression in co-culture model of Caco-2 and peripheral blood mononuclear cells (PBMC). Microb Pathog 2020; 149:104566. [DOI: 10.1016/j.micpath.2020.104566] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/01/2020] [Accepted: 10/01/2020] [Indexed: 10/23/2022]
|
22
|
Vibrio cholerae Type VI Activity Alters Motility Behavior in Mucin. J Bacteriol 2020; 202:JB.00261-20. [PMID: 32868403 DOI: 10.1128/jb.00261-20] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 08/11/2020] [Indexed: 01/16/2023] Open
Abstract
Motility is required for many bacterial pathogens to reach and colonize target sites. Vibrio cholerae traverses a thick mucus barrier coating the small intestine to reach the underlying epithelium. We screened a transposon library in motility medium containing mucin to identify factors that influence mucus transit. Lesions in structural genes of the type VI secretion system (T6SS) were among those recovered. Two-dimensional (2D) and 3D single-cell tracking was used to compare the motility behaviors of wild-type cells and a mutant that collectively lacked three essential T6SS structural genes (T6SS-). In the absence of mucin, wild-type and T6SS- cells exhibited similar speeds and run-reverse-flick (RRF) swimming patterns, in which forward-moving cells briefly backtrack before stochastically reorienting (flicking) in a new direction upon resuming forward movement. We show that mucin induced T6SS expression and activity in wild-type bacteria but significantly decreased their swimming speed and flicking, yielding curvilinear or near-surface circular traces for many cells. Conversely, mucin slowed T6SS- cells to a lesser extent, and many continued to flick and produce RRF-like traces. ΔcheY3 cells, which exclusively swim in the forward direction and thus cannot flick, also produced curvilinear traces with or without mucin present and, on occasion, near-surface circular traces in the presence of mucin. The dependence of flicking on swimming speed suggested that mucin-induced T6SS activity further decreased V. cholerae motility and thereby reduced flicking probability during reverse-to-forward transitions. We propose that this encourages cells to continue on their current trajectory rather than reorienting, which may benefit those tracking toward the epithelial surface.IMPORTANCE V. cholerae deploys an arsenal of virulence factors as it attempts to traverse a protective mucus layer and reach the epithelial surface of the distal small intestine. The T6SS used to cull bacterial competition during infection is induced by mucus. We show that this activity may serve an additional purpose by further decreasing motility in the presence of mucin, thereby reducing the probability of speed-dependent, near-perpendicular directional changes. We posit that this encourages cells to maintain course rather than change direction, which may aid those attempting to reach and colonize the epithelial surface.
Collapse
|
23
|
Abstract
Vibrio cholerae remains a challenge in the developing world and incidence of the disease it causes, cholera, is anticipated to increase with rising global temperatures and with emergent, highly infectious strains. At present, the underlying metabolic processes that support V. cholerae growth during infection are less well understood than specific virulence traits, such as production of a toxin or pilus. In this study, we determined that oxidative metabolism of host substrates such as mucin contribute significantly to V. cholerae population expansion in vivo. Identifying metabolic pathways critical for growth can provide avenues for controlling V. cholerae infection and the knowledge may be translatable to other pathogens of the gastrointestinal tract. Vibrio cholerae replicates to high cell density in the human small intestine, leading to the diarrheal disease cholera. During infection, V. cholerae senses and responds to environmental signals that govern cellular responses. Spatial localization of V. cholerae within the intestine affects nutrient availability and metabolic pathways required for replicative success. Metabolic processes used by V. cholerae to reach such high cell densities are not fully known. We sought to better define the metabolic traits that contribute to high levels of V. cholerae during infection. By disrupting the pyruvate dehydrogenase (PDH) complex and pyruvate formate-lyase (PFL), we could differentiate aerobic and anaerobic metabolic pathway involvement in V. cholerae proliferation. We demonstrate that oxidative metabolism is a key contributor to the replicative success of V. choleraein vivo using an infant mouse model in which PDH mutants were attenuated 100-fold relative to the wild type for colonization. Additionally, metabolism of host substrates, including mucin, was determined to support V. cholerae growth in vitro as a sole carbon source, primarily under aerobic growth conditions. Mucin likely contributes to population expansion during human infection as it is a ubiquitous source of carbohydrates. These data highlight oxidative metabolism as important in the intestinal environment and warrant further investigation of how oxygen and other host substrates shape the intestinal landscape that ultimately influences bacterial disease. We conclude from our results that oxidative metabolism of host substrates is a key driver of V. cholerae proliferation during infection, leading to the substantial bacterial burden exhibited in cholera patients.
Collapse
|
24
|
Spatiotemporal Regulation of Vibrio Exotoxins by HlyU and Other Transcriptional Regulators. Toxins (Basel) 2020; 12:toxins12090544. [PMID: 32842612 PMCID: PMC7551375 DOI: 10.3390/toxins12090544] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 08/17/2020] [Accepted: 08/19/2020] [Indexed: 02/06/2023] Open
Abstract
After invading a host, bacterial pathogens secrete diverse protein toxins to disrupt host defense systems. To ensure successful infection, however, pathogens must precisely regulate the expression of those exotoxins because uncontrolled toxin production squanders energy. Furthermore, inappropriate toxin secretion can trigger host immune responses that are detrimental to the invading pathogens. Therefore, bacterial pathogens use diverse transcriptional regulators to accurately regulate multiple exotoxin genes based on spatiotemporal conditions. This review covers three major exotoxins in pathogenic Vibrio species and their transcriptional regulation systems. When Vibrio encounters a host, genes encoding cytolysin/hemolysin, multifunctional-autoprocessing repeats-in-toxin (MARTX) toxin, and secreted phospholipases are coordinately regulated by the transcriptional regulator HlyU. At the same time, however, they are distinctly controlled by a variety of other transcriptional regulators. How this coordinated but distinct regulation of exotoxins makes Vibrio species successful pathogens? In addition, anti-virulence strategies that target the coordinating master regulator HlyU and related future research directions are discussed.
Collapse
|
25
|
Bakhshi B, Barzelighi HM, Daraei B. The anti-adhesive and anti-invasive effects of recombinant azurin on the interaction between enteric pathogens (invasive/non-invasive) and Caco-2 cells. Microb Pathog 2020; 147:104246. [PMID: 32562811 DOI: 10.1016/j.micpath.2020.104246] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 05/02/2020] [Accepted: 05/04/2020] [Indexed: 10/24/2022]
Abstract
Anti-adhesion therapy and anti-adhesin immunity are meant to diminish the interaction between pathogens and host tissues, either by prevention or by exclusion of bacterial adhesion and entrance to cells. Azurin is a scaffold protein possessing antiviral, antiparasitic, and anticancer activities. The purpose of the present study was to determine the effect of recombinant Azurin (rAzurin) on the adhesion and invasion capacity of invasive (Shigella sonnei, Shigella flexneri, Campylobacter jejuni) and non-invasive (Vibrio cholerae) enteric bacteria to cells. The non-toxic dose of rAzurin and the best MOI (Multiplicity of Infection) of bacterial species was assessed by MTT assay. Bacterial species were used at MOIs of 20:1 and Azurin was applied at the concentrations of 5 and 25 μg/mL and added to Caco-2 cells in competition and replacement assay to assess the anti-adhesion and anti-invasion properties of rAzurin. The protein caused significant decrease in the adhesion rate of S. sonnei, S. flexneri, C. jejuni, and V. cholerae strains to Caco-2 cells by 43, 39, 72, and 38% in competition and 45, 46, 75, and 48% in replacement assays, respectively. Also, S. sonnei, S. flexneri, and C. jejuni strains invasion rate was reduced to 50, 50, and 70% in anti-invasion assay, respectively. The inhibitory effect of Azurin against C. jejuni and V. cholerae strains adhesion was more significant (p < .001) compared to Shigella spp. (p < .05) which may be due to smaller size of the former bacteria. On the contrary, in invasion assay, rAzurin showed a greater inhibitory effect against Shigella spp. (p < .001) compared to C. jejuni (p < .05), which may probably be due to the interaction of rAzurin with several effectors or ligands, involved in Shigella invasion and internalization. The findings of the present study opens new insights of rAzurin as a new and potent candidate for reducing or probably preventing enteric bacterial attachment, invasion, and pathogenesis.
Collapse
Affiliation(s)
- Bita Bakhshi
- Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | | | - Bahram Daraei
- Department of Toxicology and Pharmacology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
26
|
Baranova DE, Willsey GG, Levinson KJ, Smith C, Wade J, Mantis NJ. Transcriptional profiling of Vibrio cholerae O1 following exposure to human anti- lipopolysaccharide monoclonal antibodies. Pathog Dis 2020; 78:ftaa029. [PMID: 32589220 PMCID: PMC7371154 DOI: 10.1093/femspd/ftaa029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 06/22/2020] [Indexed: 02/04/2023] Open
Abstract
Following an episode of cholera, a rapidly dehydrating, watery diarrhea caused by the Gram-negative bacterium, Vibrio cholerae O1, humans mount a robust anti-lipopolysaccharide (LPS) antibody response that is associated with immunity to subsequent re-infection. In neonatal mouse and rabbit models of cholera, passively administered anti-LPS polyclonal and monoclonal (MAb) antibodies reduce V. cholerae colonization of the intestinal epithelia by inhibiting bacterial motility and promoting vibrio agglutination. Here we demonstrate that human anti-LPS IgG MAbs also arrest V. cholerae motility and induce bacterial paralysis. A subset of those MAbs also triggered V. cholerae to secrete an extracellular matrix (ECM). To identify changes in gene expression that accompany antibody exposure and that may account for motility arrest and ECM production, we subjected V. cholerae O1 El Tor to RNA-seq analysis after treatment with ZAC-3 IgG, a high affinity MAb directed against the core/lipid A region of LPS. We identified > 160 genes whose expression was altered following ZAC-3 IgG treatment, although canonical outer membrane stress regulons were not among them. ompS (VCA1028), a porin associated with virulence and indirectly regulated by ToxT, and norR (VCA0182), a σ54-dependent transcription factor involved in late stages of infection, were two upregulated genes worth noting.
Collapse
Affiliation(s)
- Danielle E Baranova
- Department of Biomedical Sciences, University at Albany, 1400 Washington Ave, Albany NY 12222
- Division of Infectious Diseases, Wadsworth Center, NYS Department of Health, 120 New Scotland Ave, Albany NY 12208
| | - Graham G Willsey
- Division of Infectious Diseases, Wadsworth Center, NYS Department of Health, 120 New Scotland Ave, Albany NY 12208
| | - Kara J Levinson
- Department of Biomedical Sciences, University at Albany, 1400 Washington Ave, Albany NY 12222
- Division of Infectious Diseases, Wadsworth Center, NYS Department of Health, 120 New Scotland Ave, Albany NY 12208
| | - Carol Smith
- Division of Molecular Genetics, Wadsworth Center, NYS Department of Health, 120 New Scotland Ave, Albany NY 12208
| | - Joseph Wade
- Department of Biomedical Sciences, University at Albany, 1400 Washington Ave, Albany NY 12222
- Division of Molecular Genetics, Wadsworth Center, NYS Department of Health, 120 New Scotland Ave, Albany NY 12208
| | - Nicholas J Mantis
- Department of Biomedical Sciences, University at Albany, 1400 Washington Ave, Albany NY 12222
- Division of Infectious Diseases, Wadsworth Center, NYS Department of Health, 120 New Scotland Ave, Albany NY 12208
| |
Collapse
|
27
|
Baranova DE, Chen L, Destrempes M, Meade H, Mantis NJ. Passive Immunity to Vibrio cholerae O1 Afforded by a Human Monoclonal IgA1 Antibody Expressed in Milk. Pathog Immun 2020; 5:89-116. [PMID: 34136728 PMCID: PMC8204294 DOI: 10.20411/pai.v5i1.370] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 04/14/2020] [Indexed: 12/14/2022] Open
Abstract
Background: In cholera epidemics, the spread of disease can easily outpace vaccine
control measures. The advent of technologies enabling the expression of
recombinant proteins, including antibodies, in the milk of transgenic
animals raises the prospect of developing a self-administered and
cost-effective monoclonal antibody (MAb)-based prophylactic to reduce the
incidence of Vibrio cholerae infection. Methods: We generated a transgenic mouse line in which the heavy and light chain
variable regions (Fv) specific for a conserved epitope in the core/lipid A
of V. cholerae O1 lipopolysaccharide were expressed as a
full-length human dimeric IgA1 (ZAC-3) and secreted into the milk of
lactating dams. Milk containing ZAC-3 IgA1 was assessed for the ability to
passively protect against experimental cholera infection in a newborn mouse
model and to impact bacterial swimming behavior. Results: Newborn mice that were passively administered ZAC-3 IgA1 containing milk, or
that suckled on dams expressing ZAC-3 IgA1, were immune to experimental
cholera infection, as measured by a reduction of V.
cholerae O1 colony forming units recovered from intestinal
lysates 12 hours after oral challenge. In vitro analysis
revealed that ZAC-3 hIgA1-containing milk arrested V.
cholerae motility in soft agar and liquid media and was
effective at promoting bacterial agglutination, possibly accounting for the
observed reduction in bacterial colonization in vivo. Conclusions: These results demonstrate that consumption of milk-derived antibodies may
serve as a strategy to passively protect against cholera and possibly other
enteric pathogens.
Collapse
Affiliation(s)
- Danielle E Baranova
- Department of Biomedical Sciences; University at Albany; Albany, New York.,Division of Infectious Diseases; Wadsworth Center; New York State Department of Health; Albany, New York
| | | | | | | | - Nicholas J Mantis
- Department of Biomedical Sciences; University at Albany; Albany, New York.,Division of Infectious Diseases; Wadsworth Center; New York State Department of Health; Albany, New York
| |
Collapse
|
28
|
You JS, Yong JH, Kim GH, Moon S, Nam KT, Ryu JH, Yoon MY, Yoon SS. Commensal-derived metabolites govern Vibrio cholerae pathogenesis in host intestine. MICROBIOME 2019; 7:132. [PMID: 31521198 PMCID: PMC6744661 DOI: 10.1186/s40168-019-0746-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 09/03/2019] [Indexed: 05/15/2023]
Abstract
BACKGROUND Recent evidence suggests that the commensal microbes act as a barrier against invading pathogens and enteric infections are the consequences of multi-layered interactions among commensals, pathogens, and the host intestinal tissue. However, it remains unclear how perturbations of the gut microbiota compromise host infection resistance, especially through changes at species and metabolite levels. RESULTS Here, we illustrate how Bacteroides vulgatus, a dominant species of the Bacteroidetes phylum in mouse intestine, suppresses infection by Vibrio cholerae, an important human pathogen. Clindamycin (CL) is an antibiotic that selectively kills anaerobic bacteria, and accordingly Bacteroidetes are completely eradicated from CL-treated mouse intestines. The Bacteroidetes-depleted adult mice developed severe cholera-like symptoms, when infected with V. cholerae. Germ-free mice mono-associated with B. vulgatus became resistant to V. cholerae infection. Levels of V. cholerae growth-inhibitory metabolites including short-chain fatty acids plummeted upon CL treatment, while levels of compounds that enhance V. cholerae proliferation were elevated. Furthermore, the intestinal colonization process of V. cholerae was well-simulated in CL-treated adult mice. CONCLUSIONS Overall, we provide insights into how a symbiotic microbe and a pathogenic intruder interact inside host intestine. We identified B. vulgatus as an indigenous microbial species that can suppress intestinal infection. Our results also demonstrate that commensal-derived metabolites are a critical determinant for host resistance against V. cholerae infection, and that CL pretreatment of adult mice generates a simple yet useful model of cholera infection.
Collapse
Affiliation(s)
- Jin Sun You
- Department of Microbiology and Immunology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu Seoul, Seoul, 03722, Korea
- Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Ji Hyun Yong
- Department of Microbiology and Immunology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu Seoul, Seoul, 03722, Korea
- Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Gwang Hee Kim
- Department of Microbiology and Immunology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu Seoul, Seoul, 03722, Korea
- Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Sungmin Moon
- Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, 03722, Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Ki Taek Nam
- Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, 03722, Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Ji Hwan Ryu
- Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, 03722, Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Mi Young Yoon
- Department of Microbiology and Immunology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu Seoul, Seoul, 03722, Korea.
- Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, 03722, Korea.
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, Korea.
| | - Sang Sun Yoon
- Department of Microbiology and Immunology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu Seoul, Seoul, 03722, Korea.
- Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, 03722, Korea.
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, Korea.
| |
Collapse
|
29
|
DNA-uptake pili of Vibrio cholerae are required for chitin colonization and capable of kin recognition via sequence-specific self-interaction. Nat Microbiol 2019; 4:1545-1557. [PMID: 31182799 PMCID: PMC6708440 DOI: 10.1038/s41564-019-0479-5] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 05/01/2019] [Indexed: 12/02/2022]
Abstract
How bacteria colonise surfaces and how they distinguish the individuals around them are fundamental biological questions. Type IV pili are a widespread and multi-purpose class of cell surface polymers. Here we directly visualise the DNA-uptake pilus of Vibrio cholerae, which is produced specifically during growth upon its natural habitat - chitinous surfaces. As predicted, these pili are highly dynamic and retract prior to DNA-uptake during competence for natural transformation. Interestingly, DNA-uptake pili can also self-interact to mediate auto-aggregation. This capability is conserved in disease-causing pandemic strains, which typically encode the same major pilin subunit, PilA. Unexpectedly, however, we discovered that extensive strain-to-strain variability in PilA, present in environmental isolates, creates a set of highly specific interactions, enabling cells producing pili composed of different PilA subunits to distinguish between one another. We go on to show that DNA-uptake pili bind to chitinous surfaces, are required for chitin colonisation under flow, and that pili capable of self-interaction connect cells on chitin within dense pili networks. Our results suggest a model whereby DNA-uptake pili function to promote inter-bacterial interactions during surface colonisation. Moreover, they provide evidence that type IV pili could offer a simple and potentially widespread mechanism for bacterial kin recognition.
Collapse
|
30
|
Taylor VL, Fitzpatrick AD, Islam Z, Maxwell KL. The Diverse Impacts of Phage Morons on Bacterial Fitness and Virulence. Adv Virus Res 2019; 103:1-31. [PMID: 30635074 DOI: 10.1016/bs.aivir.2018.08.001] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The viruses that infect bacteria, known as phages, are the most abundant biological entity on earth. They play critical roles in controlling bacterial populations through phage-mediated killing, as well as through formation of bacterial lysogens. In this form, the survival of the phage depends on the survival of the bacterial host in which it resides. Thus, it is advantageous for phages to encode genes that contribute to bacterial fitness and expand the environmental niche. In many cases, these fitness factors also make the bacteria better able to survive in human infections and are thereby considered pathogenesis or virulence factors. The genes that encode these fitness factors, known as "morons," have been shown to increase bacterial fitness through a wide range of mechanisms and play important roles in bacterial diseases. This review outlines the benefits provided by phage morons in various aspects of bacterial life, including phage and antibiotic resistance, motility, adhesion and quorum sensing.
Collapse
Affiliation(s)
| | | | - Zafrin Islam
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Karen L Maxwell
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
31
|
Charles-Orszag A, Tsai FC, Bonazzi D, Manriquez V, Sachse M, Mallet A, Salles A, Melican K, Staneva R, Bertin A, Millien C, Goussard S, Lafaye P, Shorte S, Piel M, Krijnse-Locker J, Brochard-Wyart F, Bassereau P, Duménil G. Adhesion to nanofibers drives cell membrane remodeling through one-dimensional wetting. Nat Commun 2018; 9:4450. [PMID: 30361638 PMCID: PMC6202395 DOI: 10.1038/s41467-018-06948-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 09/19/2018] [Indexed: 01/22/2023] Open
Abstract
The shape of cellular membranes is highly regulated by a set of conserved mechanisms that can be manipulated by bacterial pathogens to infect cells. Remodeling of the plasma membrane of endothelial cells by the bacterium Neisseria meningitidis is thought to be essential during the blood phase of meningococcal infection, but the underlying mechanisms are unclear. Here we show that plasma membrane remodeling occurs independently of F-actin, along meningococcal type IV pili fibers, by a physical mechanism that we term 'one-dimensional' membrane wetting. We provide a theoretical model that describes the physical basis of one-dimensional wetting and show that this mechanism occurs in model membranes interacting with nanofibers, and in human cells interacting with extracellular matrix meshworks. We propose one-dimensional wetting as a new general principle driving the interaction of cells with their environment at the nanoscale that is diverted by meningococci during infection.
Collapse
Affiliation(s)
- Arthur Charles-Orszag
- Pathogenesis of Vascular Infections Unit, INSERM, Institut Pasteur, Paris, 75015, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, 75006, France
| | - Feng-Ching Tsai
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS UMR168, Paris, 75005, France.,Sorbonne Université, Paris, 75005, France
| | - Daria Bonazzi
- Pathogenesis of Vascular Infections Unit, INSERM, Institut Pasteur, Paris, 75015, France
| | - Valeria Manriquez
- Pathogenesis of Vascular Infections Unit, INSERM, Institut Pasteur, Paris, 75015, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, 75006, France
| | | | | | | | - Keira Melican
- Pathogenesis of Vascular Infections Unit, INSERM, Institut Pasteur, Paris, 75015, France.,Department of Neuroscience, Swedish Medical Nanoscience Center, Karolinska Institutet, Solna, 171 77, Sweden
| | - Ralitza Staneva
- Institut Curie, PSL Research University, CNRS, UMR 144, Paris, 75005, France
| | - Aurélie Bertin
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS UMR168, Paris, 75005, France.,Sorbonne Université, Paris, 75005, France
| | | | - Sylvie Goussard
- Pathogenesis of Vascular Infections Unit, INSERM, Institut Pasteur, Paris, 75015, France
| | - Pierre Lafaye
- Antibody Engineering, Institut Pasteur, Paris, 75015, France
| | | | - Matthieu Piel
- Systems Biology of Cell Polarity and Cell Division, Institut Pierre-Gilles De Gennes, Paris, 75005, France.,Institut Curie, Paris, 75005, France
| | | | - Françoise Brochard-Wyart
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS UMR168, Paris, 75005, France.,Sorbonne Université, Paris, 75005, France
| | - Patricia Bassereau
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS UMR168, Paris, 75005, France.,Sorbonne Université, Paris, 75005, France
| | - Guillaume Duménil
- Pathogenesis of Vascular Infections Unit, INSERM, Institut Pasteur, Paris, 75015, France.
| |
Collapse
|
32
|
Kim HY, Go J, Lee KM, Oh YT, Yoon SS. Guanosine tetra- and pentaphosphate increase antibiotic tolerance by reducing reactive oxygen species production in Vibrio cholerae. J Biol Chem 2018; 293:5679-5694. [PMID: 29475943 PMCID: PMC5900777 DOI: 10.1074/jbc.ra117.000383] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 02/16/2018] [Indexed: 02/06/2023] Open
Abstract
The pathogen Vibrio cholerae is the causative agent of cholera. Emergence of antibiotic-resistant V. cholerae strains is increasing, but the underlying mechanisms remain unclear. Herein, we report that the stringent response regulator and stress alarmone guanosine tetra- and pentaphosphate ((p)ppGpp) significantly contributes to antibiotic tolerance in V. cholerae We found that N16961, a pandemic V. cholerae strain, and its isogenic (p)ppGpp-overexpressing mutant ΔrelAΔspoT are both more antibiotic-resistant than (p)ppGpp0 (ΔrelAΔrelVΔspoT) and ΔdksA mutants, which cannot produce or utilize (p)ppGpp, respectively. We also found that additional disruption of the aconitase B-encoding and tricarboxylic acid (TCA) cycle gene acnB in the (p)ppGpp0 mutant increases its antibiotic tolerance. Moreover, expression of TCA cycle genes, including acnB, was increased in (p)ppGpp0, but not in the antibiotic-resistant ΔrelAΔspoT mutant, suggesting that (p)ppGpp suppresses TCA cycle activity, thereby entailing antibiotic resistance. Importantly, when grown anaerobically or incubated with an iron chelator, the (p)ppGpp0 mutant became antibiotic-tolerant, suggesting that reactive oxygen species (ROS) are involved in antibiotic-mediated bacterial killing. Consistent with that hypothesis, tetracycline treatment markedly increased ROS production in the antibiotic-susceptible mutants. Interestingly, expression of the Fe(III) ABC transporter substrate-binding protein FbpA was increased 10-fold in (p)ppGpp0, and fbpA gene deletion restored viability of tetracycline-exposed (p)ppGpp0 cells. Of note, FbpA expression was repressed in the (p)ppGpp-accumulating mutant, resulting in a reduction of intracellular free iron, required for the ROS-generating Fenton reaction. Our results indicate that (p)ppGpp-mediated suppression of central metabolism and iron uptake reduces antibiotic-induced oxidative stress in V. cholerae.
Collapse
Affiliation(s)
- Hwa Young Kim
- From the Department of Microbiology and Immunology, Brain Korea 21 PLUS Project for Medical Science, and ,the Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Korea and
| | - Junhyeok Go
- From the Department of Microbiology and Immunology, Brain Korea 21 PLUS Project for Medical Science, and ,the Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Korea and
| | - Kang-Mu Lee
- From the Department of Microbiology and Immunology, Brain Korea 21 PLUS Project for Medical Science, and ,the Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Korea and
| | - Young Taek Oh
- From the Department of Microbiology and Immunology, Brain Korea 21 PLUS Project for Medical Science, and ,the Freshwater Bioresources Utilization Division, Nakdonggang National Institute of Biological Resources, SangJu 37242, Korea, To whom correspondence may be addressed:
Freshwater Bioresources Utilization Division, Nakdonggang National Institute of Biological Resources, SangJu-si 37242, Korea. Tel.:
82-54-530-0932; Fax:
82-54-530-0949; E-mail:
| | - Sang Sun Yoon
- From the Department of Microbiology and Immunology, Brain Korea 21 PLUS Project for Medical Science, and ,the Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Korea and , To whom correspondence may be addressed:
Dept. of Microbiology and Immunology, Yonsei University College of Medicine, 250 Seongsanno, Seodaemun-gu Seoul 120-752, Korea. Tel.:
82-2-2228-1824; Fax:
82-2-392-7088; E-mail:
| |
Collapse
|
33
|
Fu Y, Ho BT, Mekalanos JJ. Tracking Vibrio cholerae Cell-Cell Interactions during Infection Reveals Bacterial Population Dynamics within Intestinal Microenvironments. Cell Host Microbe 2018; 23:274-281.e2. [PMID: 29398650 PMCID: PMC6031135 DOI: 10.1016/j.chom.2017.12.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 11/09/2017] [Accepted: 12/13/2017] [Indexed: 01/20/2023]
Abstract
Vibrio cholerae is the causative agent of the diarrheal disease cholera. Although many V. cholerae virulence factors have been studied, the role of interbacterial interactions within the host gut and their influence on colonization are poorly understood. Here, we utilized the conjugative properties of a Vibrio-specific plasmid to serve as a quantifiable genetic marker for direct contact among V. cholerae cells in the infant rabbit model for cholera. In conjunction, we also quantified contact-dependent type 6 secretion system (T6SS)-mediated killing of co-infecting V. cholerae strains. Tracking these interbacterial interactions revealed that most contact-dependent cell-cell interactions among V. cholerae occur in specific intestinal microenvironments, notably the distal small intestine and cecum, and that the T6SS confers a competitive advantage within the middle small intestine. These results support a model for V. cholerae gut colonization, which includes microenvironments where critical microbial-host and bacterial-bacterial interactions occur to facilitate colonization by this pathogen.
Collapse
Affiliation(s)
- Yang Fu
- Department of Microbiology and Immunobiology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Brian T Ho
- Department of Microbiology and Immunobiology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - John J Mekalanos
- Department of Microbiology and Immunobiology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.
| |
Collapse
|
34
|
Begum YA, Rydberg HA, Thorell K, Kwak YK, Sun L, Joffré E, Qadri F, Sjöling Å. In Situ Analyses Directly in Diarrheal Stool Reveal Large Variations in Bacterial Load and Active Toxin Expression of Enterotoxigenic Escherichiacoli and Vibrio cholerae. mSphere 2018; 3:e00517-17. [PMID: 29404412 PMCID: PMC5784243 DOI: 10.1128/msphere.00517-17] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 12/27/2017] [Indexed: 11/20/2022] Open
Abstract
The bacterial pathogens enterotoxigenic Escherichia coli (ETEC) and Vibrio cholerae are major causes of diarrhea. ETEC causes diarrhea by production of the heat-labile toxin (LT) and heat-stable toxins (STh and STp), while V. cholerae produces cholera toxin (CT). In this study, we determined the occurrence and bacterial doses of the two pathogens and their respective toxin expression levels directly in liquid diarrheal stools of patients in Dhaka, Bangladesh. By quantitative culture and real-time quantitative PCR (qPCR) detection of the toxin genes, the two pathogens were found to coexist in several of the patients, at concentrations between 102 and 108 bacterial gene copies per ml. Even in culture-negative samples, gene copy numbers of 102 to 104 of either ETEC or V. cholerae toxin genes were detected by qPCR. RNA was extracted directly from stool, and gene expression levels, quantified by reverse transcriptase qPCR (RT-qPCR), of the genes encoding CT, LT, STh, and STp showed expression of toxin genes. Toxin enzyme-linked immunosorbent assay (ELISA) confirmed active toxin secretion directly in the liquid diarrhea. Analysis of ETEC isolates by multiplex PCR, dot blot analysis, and genome sequencing suggested that there are genetic ETEC profiles that are more commonly found as dominating single pathogens and others that are coinfectants with lower bacterial loads. The ETEC genomes, including assembled genomes of dominating ETEC isolates expressing LT/STh/CS5/CS6 and LT/CS7, are provided. In addition, this study highlights an emerging important ETEC strain expressing LT/STp and the novel colonization factor CS27b. These findings have implications for investigations of pathogenesis as well as for vaccine development. IMPORTANCE The cause of diarrheal disease is usually determined by screening for several microorganisms by various methods, and sole detection is used to assign the agent as the cause of disease. However, it has become increasingly clear that many infections are caused by coinfections with several pathogens and that the dose of the infecting pathogen is important. We quantified the absolute numbers of enterotoxigenic E. coli (ETEC) and Vibrio cholerae directly in diarrheal fluid. We noted several events where both pathogens were found but also a large dose dependency. In three samples, we found ETEC as the only pathogen sought for. These isolates belonged to globally distributed ETEC clones and were the dominating species in stool with active toxin expression. This suggests that certain superior virulent ETEC lineages are able to outcompete the gut microbiota and be the sole cause of disease and hence need to be specifically monitored.
Collapse
Affiliation(s)
- Yasmin Ara Begum
- International Centre for Diarrhoeal Disease Research, Bangladesh, Centre for Health and Population Research, Dhaka, Bangladesh
| | - Hanna A. Rydberg
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research (CTMR), Karolinska Institutet, Stockholm, Sweden
| | - Kaisa Thorell
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research (CTMR), Karolinska Institutet, Stockholm, Sweden
| | - Young-Keun Kwak
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research (CTMR), Karolinska Institutet, Stockholm, Sweden
| | - Lei Sun
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research (CTMR), Karolinska Institutet, Stockholm, Sweden
| | - Enrique Joffré
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research (CTMR), Karolinska Institutet, Stockholm, Sweden
| | - Firdausi Qadri
- International Centre for Diarrhoeal Disease Research, Bangladesh, Centre for Health and Population Research, Dhaka, Bangladesh
| | - Åsa Sjöling
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research (CTMR), Karolinska Institutet, Stockholm, Sweden
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
35
|
|
36
|
Mewborn L, Benitez JA, Silva AJ. Flagellar motility, extracellular proteases and Vibrio cholerae detachment from abiotic and biotic surfaces. Microb Pathog 2017; 113:17-24. [PMID: 29038053 DOI: 10.1016/j.micpath.2017.10.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 10/09/2017] [Accepted: 10/12/2017] [Indexed: 12/21/2022]
Abstract
Vibrio cholerae of serogroups O1 and O139, the causative agent of Asiatic cholera, continues to be a major global health threat. This pathogen utilizes substratum-specific pili to attach to distinct surfaces in the aquatic environment and the human small intestine and detaches when conditions become unfavorable. Both attachment and detachment are critical to bacterial environmental survival, pathogenesis and disease transmission. However, the factors that promote detachment are less understood. In this study, we examine the role of flagellar motility and hemagglutinin/protease (HapA) in vibrio detachment from a non-degradable abiotic surface and from the suckling mouse intestine. Flagellar motility facilitated V. cholerae detachment from abiotic surfaces. HapA had no effect on the stability of biofilms formed on abiotic surfaces despite representing >50% of the proteolytic activity present in the extracellular matrix. We developed a balanced lethal plasmid system to increase the bacterial cyclic diguanylate (c-di-GMP) pool late in infection, a condition that represses motility and HapA expression. Increasing the c-di-GMP pool enhanced V. cholerae colonization of the suckling mouse intestine. The c-di-GMP effect was fully abolished in hapA isogenic mutants. These results suggest that motility facilitates detachment in a substratum-independent manner. Instead, HapA appears to function as a substratum-specific detachment factor.
Collapse
Affiliation(s)
- Loree Mewborn
- Morehouse School of Medicine, Department of Microbiology, Biochemistry and Immunology, 720 Westview Dr., SW Atlanta, 30310, GA, USA
| | - Jorge A Benitez
- Morehouse School of Medicine, Department of Microbiology, Biochemistry and Immunology, 720 Westview Dr., SW Atlanta, 30310, GA, USA
| | - Anisia J Silva
- Morehouse School of Medicine, Department of Microbiology, Biochemistry and Immunology, 720 Westview Dr., SW Atlanta, 30310, GA, USA.
| |
Collapse
|
37
|
Bhattaram V, Upadhyay A, Yin HB, Mooyottu S, Venkitanarayanan K. Effect of Dietary Minerals on Virulence Attributes of Vibrio cholerae. Front Microbiol 2017; 8:911. [PMID: 28579983 PMCID: PMC5437166 DOI: 10.3389/fmicb.2017.00911] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Accepted: 05/04/2017] [Indexed: 12/22/2022] Open
Abstract
Vibrio cholerae is a water-borne pathogen responsible for causing a toxin-mediated profuse diarrhea in humans, leading to severe dehydration and death in unattended patients. With increasing reports of antibiotic resistance in V. cholerae, there is a need for alternate interventional strategies for controlling cholera. A potential new strategy for treating infectious diseases involves targeting bacterial virulence rather than growth, where a pathogen’s specific mechanisms critical for causing infection in hosts are inhibited. Since bacterial motility, intestinal colonization and cholera toxin are critical components in V. cholerae pathogenesis, attenuating these virulence factors could potentially control cholera in humans. In this study, the efficacy of sub-inhibitory concentration (SIC, highest concentration not inhibiting bacterial growth) of essential minerals, zinc (Zn), selenium (Se), and manganese (Mn) in reducing V. cholerae motility and adhesion to intestinal epithelial cells (Caco-2), cholera toxin production, and toxin binding to the ganglioside receptor (GM1) was investigated. Additionally, V. cholerae attachment and toxin production in an ex vivo mouse intestine model was determined. Further, the effect of Zn, Se and Mn on V. cholerae virulence genes, ctxAB (toxin production), fliA (motility), tcpA (intestinal colonization), and toxR (master regulon) was determined using real-time quantitative PCR. All three minerals significantly reduced V. cholerae motility, adhesion to Caco-2 cells, and cholera toxin production in vitro, and decreased adhesion and toxin production in mouse intestine ex vivo (P < 0.05). In addition, Zn, Se, and Mn down-regulated the transcription of virulence genes, ctxAB, fliA, and toxR. Results suggest that Zn, Se, and Mn could be potentially used to reduce V. cholerae virulence. However, in vivo studies in an animal model are necessary to validate these results.
Collapse
Affiliation(s)
- Varunkumar Bhattaram
- Department of Animal Science, University of Connecticut, StorrsCT, United States
| | - Abhinav Upadhyay
- Department of Animal Science, University of Connecticut, StorrsCT, United States.,Department of Poultry Science, University of Arkansas, FayettevilleAR, United States
| | - Hsin-Bai Yin
- Department of Animal Science, University of Connecticut, StorrsCT, United States
| | - Shankumar Mooyottu
- Department of Animal Science, University of Connecticut, StorrsCT, United States
| | | |
Collapse
|
38
|
A new class of inhibitors of the AraC family virulence regulator Vibrio cholerae ToxT. Sci Rep 2017; 7:45011. [PMID: 28332578 PMCID: PMC5362913 DOI: 10.1038/srep45011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 02/20/2017] [Indexed: 11/28/2022] Open
Abstract
Vibrio cholerae is responsible for the diarrheal disease cholera that infects millions of people worldwide. While vaccines protecting against cholera exist, and oral rehydration therapy is an effective treatment method, the disease will remain a global health threat until long-term solutions such as improved sanitation and access to clean water become widely available. Because of this, there is a pressing need for potent therapeutics that can either mitigate cholera symptoms, or act prophylactically to prevent the virulent effects of a cholera infection. Here we report the design, synthesis, and characterization of a set of compounds that bind and inhibit ToxT, the transcription factor that directly regulates the two primary V. cholerae virulence factors. Using the folded structure of the monounsaturated fatty acid observed in the X-ray structure of ToxT as a template, we designed ten novel compounds that inhibit the virulence cascade to a greater degree than any known inhibitor. Our findings provide a structural and functional basis for the development of viable antivirulence therapeutics that combat cholera and, potentially, other forms of bacterial pathogenic disease.
Collapse
|
39
|
Nithichanon A, Gourlay LJ, Bancroft GJ, Ato M, Takahashi Y, Lertmemongkolchai G. Boosting of post-exposure human T-cell and B-cell recall responses in vivo by Burkholderia pseudomallei-related proteins. Immunology 2017; 151:98-109. [PMID: 28066900 DOI: 10.1111/imm.12709] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Revised: 12/30/2016] [Accepted: 01/04/2017] [Indexed: 12/20/2022] Open
Abstract
Burkholderia pseudomallei is the causative agent of melioidosis, an infectious disease with high incidence and mortality in South East Asia and northern Australia. To date there is no protective vaccine and antibiotic treatment is prolonged and not always effective. Most people living in endemic areas have been exposed to the bacteria and have developed some immunity, which may have helped to prevent disease. Here, we used a humanized mouse model (hu-PBL-SCID), reconstituted with human peripheral blood mononuclear cells from seropositive donors, to illustrate the potential of three known antigens (FliC, OmpA and N-PilO2) for boosting both T-cell and B-cell immune responses. All three antigens boosted the production of specific antibodies in vivo, and increased the number of antibody and interferon-γ-secreting cells, and induced antibody affinity maturation. Moreover, antigen-specific antibodies isolated from either seropositive individuals or boosted mice, were found to enhance phagocytosis and oxidative burst activities from human polymorphonuclear cells. Our study demonstrates that FliC, OmpA and N-PilO2 can stimulate human memory T and B cells and highlight the potential of the hu-PBL-SCID system for screening and evaluation of novel protein antigens for inclusion in future vaccine trials against melioidosis.
Collapse
Affiliation(s)
- Arnone Nithichanon
- The Centre for Research and Development of Medical Diagnostic Laboratories, Khon Kaen University, Khon Kaen, Thailand
| | | | | | - Manabu Ato
- Department of Immunology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yoshimasa Takahashi
- Department of Immunology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Ganjana Lertmemongkolchai
- The Centre for Research and Development of Medical Diagnostic Laboratories, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
40
|
Chang YW, Kjær A, Ortega DR, Kovacikova G, Sutherland JA, Rettberg LA, Taylor RK, Jensen GJ. Architecture of the Vibrio cholerae toxin-coregulated pilus machine revealed by electron cryotomography. Nat Microbiol 2017; 2:16269. [PMID: 28165453 PMCID: PMC5302817 DOI: 10.1038/nmicrobiol.2016.269] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 12/14/2016] [Indexed: 01/22/2023]
Affiliation(s)
- Yi-Wei Chang
- California Institute of Technology, Pasadena, California 91125, USA
| | - Andreas Kjær
- University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Davi R Ortega
- California Institute of Technology, Pasadena, California 91125, USA
| | | | - John A Sutherland
- Geisel School of Medicine at Dartmouth, Hanover, New Hampshire 03755, USA
| | - Lee A Rettberg
- Howard Hughes Medical Institute, Pasadena, California 91125, USA
| | - Ronald K Taylor
- Geisel School of Medicine at Dartmouth, Hanover, New Hampshire 03755, USA
| | - Grant J Jensen
- California Institute of Technology, Pasadena, California 91125, USA.,Howard Hughes Medical Institute, Pasadena, California 91125, USA
| |
Collapse
|
41
|
The Live Attenuated Cholera Vaccine CVD 103-HgR Primes Responses to the Toxin-Coregulated Pilus Antigen TcpA in Subjects Challenged with Wild-Type Vibrio cholerae. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017; 24:CVI.00470-16. [PMID: 27847368 DOI: 10.1128/cvi.00470-16] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 11/04/2016] [Indexed: 11/20/2022]
Abstract
One potential advantage of live attenuated bacterial vaccines is the ability to stimulate responses to antigens which are only expressed during the course of infection. To determine whether the live attenuated cholera vaccine CVD 103-HgR (Vaxchora) results in antibody responses to the in vivo-induced toxin-coregulated pilus antigen TcpA, we measured IgA and IgG responses to Vibrio cholerae O1 El Tor TcpA in a subset of participants in a recently reported experimental challenge study. Participants were challenged with V. cholerae O1 El Tor Inaba N16961 either 10 days or 90 days after receiving the vaccine or a placebo. Neither vaccination nor experimental infection with V. cholerae alone resulted in a robust TcpA IgG or IgA response, but each did elicit a strong response to cholera toxin. However, compared to placebo recipients, vaccinees had a marked increase in IgG TcpA antibodies following the 90-day challenge, suggesting that vaccination with CVD 103-HgR resulted in priming for a subsequent response to TcpA. No such difference between vaccine and placebo recipients was observed for volunteers challenged 10 days after vaccination, indicating that this was insufficient time for vaccine-induced priming of the TcpA response. The priming of the response to TcpA and potentially other antigens expressed in vivo by attenuated V. cholerae may have relevance to the maintenance of immunity in areas where cholera is endemic.
Collapse
|
42
|
Abstract
Vibrio cholerae has caused seven cholera pandemics since 1817, imposing terror on much of the world, but bacterial strains are currently only available for the sixth and seventh pandemics. The El Tor biotype seventh pandemic began in 1961 in Indonesia, but did not originate directly from the classical biotype sixth-pandemic strain. Previous studies focused mainly on the spread of the seventh pandemic after 1970. Here, we analyze in unprecedented detail the origin, evolution, and transition to pandemicity of the seventh-pandemic strain. We used high-resolution comparative genomic analysis of strains collected from 1930 to 1964, covering the evolution from the first available El Tor biotype strain to the start of the seventh pandemic. We define six stages leading to the pandemic strain and reveal all key events. The seventh pandemic originated from a nonpathogenic strain in the Middle East, first observed in 1897. It subsequently underwent explosive diversification, including the spawning of the pandemic lineage. This rapid diversification suggests that, when first observed, the strain had only recently arrived in the Middle East, possibly from the Asian homeland of cholera. The lineage migrated to Makassar, Indonesia, where it gained the important virulence-associated elements Vibrio seventh pandemic island I (VSP-I), VSP-II, and El Tor type cholera toxin prophage by 1954, and it then became pandemic in 1961 after only 12 additional mutations. Our data indicate that specific niches in the Middle East and Makassar were important in generating the pandemic strain by providing gene sources and the driving forces for genetic events.
Collapse
|
43
|
Levinson KJ, Baranova DE, Mantis NJ. A monoclonal antibody that targets the conserved core/lipid A region of lipopolysaccharide affects motility and reduces intestinal colonization of both classical and El Tor Vibrio cholerae biotypes. Vaccine 2016; 34:5833-5836. [PMID: 27773473 DOI: 10.1016/j.vaccine.2016.10.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 09/26/2016] [Accepted: 10/08/2016] [Indexed: 12/20/2022]
Abstract
Vibrio cholerae is the causative agent of cholera, an acute diarrheal disease that remains endemic in many parts of the world. The mechanisms underlying immunity to cholera remain poorly defined, though it is increasingly clear that protection is associated with antibodies against lipopolysaccharide (LPS). Here we report that ZAC-3, a monoclonal antibody against the core/lipid A region of V. cholerae LPS is a potent inhibitor of V. cholerae flagellum-based motility in viscous and liquid environments. ZAC-3 arrested motility of the classical Ogawa strain O395, as well as the El Tor Inaba strain C6706. In addition, we demonstrate, in the neonatal mouse model, that ZAC-3 IgG and Fab fragments significantly reduced the ability of both V. cholerae strains O395 and C6706 to colonize the intestinal epithelium, revealing the potential of antibodies against the core/lipid A to contribute to immunity across biotypes, possibly through a mechanism involving motility arrest.
Collapse
Affiliation(s)
- Kara J Levinson
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY 12208, United States; Department of Biomedical Sciences, University at Albany, Albany, NY 12208, United States
| | - Danielle E Baranova
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY 12208, United States; Department of Biomedical Sciences, University at Albany, Albany, NY 12208, United States
| | - Nicholas J Mantis
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY 12208, United States; Department of Biomedical Sciences, University at Albany, Albany, NY 12208, United States.
| |
Collapse
|
44
|
Foulon V, Le Roux F, Lambert C, Huvet A, Soudant P, Paul-Pont I. Colonization of Polystyrene Microparticles by Vibrio crassostreae: Light and Electron Microscopic Investigation. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2016; 50:10988-10996. [PMID: 27640445 DOI: 10.1021/acs.est.6b02720] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Microplastics collected at sea harbor a high diversity of microorganisms, including some Vibrio genus members, raising questions about the role of microplastics as a novel ecological niche for potentially pathogenic microorganisms. In the present study, we investigated the adhesion dynamics of Vibrio crassostreae on polystyrene microparticles (micro-PS) using electronic and fluorescence microscopy techniques. Micro-PS were incubated with bacteria in different media (Zobell culture medium and artificial seawater) with or without natural marine aggregates. The highest percentage of colonized particles (38-100%) was observed in Zobell culture medium, which may be related to nutrient availability for production of pili and exopolysaccharide adhesion structures. A longer bacterial attachment (6 days) was observed on irregular micro-PS compared to smooth particles (<10 h), but complete decolonization of all particles eventually occurred. The presence of natural marine agreggates around micro-PS led to substantial and perennial colonization featuring monospecific biofilms at the surface of the aggregates. These exploratory results suggest that V. crassostreae may be a secondary colonizer of micro-PS, requiring a multispecies community to form a durable adhesion phenotype. Temporal assessment of microbial colonization on microplastics at sea using imaging and omics approaches are further indicated to better understand the microplastics colonization dynamics and species assemblages.
Collapse
Affiliation(s)
- Valentin Foulon
- Laboratoire des Sciences de l'Environnement Marin (LEMAR), UMR 6539 CNRS UBO IRD Ifremer, Institut Universitaire Européen de la Mer, Technopôle Brest-Iroise , Rue Dumont d'Urville, 29280 Plouzané, France
| | - Frédérique Le Roux
- Ifremer, Unité Physiologie Fonctionnelle des Organismes Marins , ZI de la Pointe du Diable, CS 10070, F-29280 Plouzané, France
- Sorbonne Universités, UPMC Paris 06, CNRS, UMR 8227, Integrative Biology of Marine Models , Station Biologique de Roscoff, CS 90074, F-29688 Roscoff, France
| | - Christophe Lambert
- Laboratoire des Sciences de l'Environnement Marin (LEMAR), UMR 6539 CNRS UBO IRD Ifremer, Institut Universitaire Européen de la Mer, Technopôle Brest-Iroise , Rue Dumont d'Urville, 29280 Plouzané, France
| | - Arnaud Huvet
- Ifremer, Laboratoire des Sciences de l'Environnement Marin (LEMAR, UMR 6539 UBO/CNRS/IRD/Ifremer), Centre Bretagne , ZI de la Pointe du Diable, CS 10070, 29280 Plouzané, France
| | - Philippe Soudant
- Laboratoire des Sciences de l'Environnement Marin (LEMAR), UMR 6539 CNRS UBO IRD Ifremer, Institut Universitaire Européen de la Mer, Technopôle Brest-Iroise , Rue Dumont d'Urville, 29280 Plouzané, France
| | - Ika Paul-Pont
- Laboratoire des Sciences de l'Environnement Marin (LEMAR), UMR 6539 CNRS UBO IRD Ifremer, Institut Universitaire Européen de la Mer, Technopôle Brest-Iroise , Rue Dumont d'Urville, 29280 Plouzané, France
| |
Collapse
|
45
|
Single Nucleotide Polymorphisms in Regulator-Encoding Genes Have an Additive Effect on Virulence Gene Expression in a Vibrio cholerae Clinical Isolate. mSphere 2016; 1:mSphere00253-16. [PMID: 27668288 PMCID: PMC5031793 DOI: 10.1128/msphere.00253-16] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 09/02/2016] [Indexed: 11/21/2022] Open
Abstract
Cholera, an infectious disease of the small intestine caused by the aquatic bacterium Vibrio cholerae, often results in vomiting and acute watery diarrhea. If left untreated or if the response is too slow, the symptoms can quickly lead to extreme dehydration and ultimately death of the patient. Recent anecdotal evidence of cholera patients suffering from increasingly severe symptoms and of disease progression at a much higher rate than previously observed has emerged. As recent cholera outbreaks caused by increasingly virulent strains have resulted in higher mortality rates, the need to investigate the mechanism(s) allowing this observed increased virulence is apparent. The significance of our research is in identifying the mechanism for increased virulence capabilities, which will allow the development of a model that will greatly enhance our understanding of cholera disease and V. cholerae pathogenesis, leading to broader biomedical impacts, as cholera serves as a model for other enteric diarrheal diseases. Vibrio cholerae is the etiological agent of the infectious disease cholera, which is characterized by vomiting and severe watery diarrhea. Recently, V. cholerae clinical isolates have demonstrated increased virulence capabilities, causing more severe symptoms with a much higher rate of disease progression than previously observed. We have identified single nucleotide polymorphisms (SNPs) in four virulence-regulatory genes (hapR, hns, luxO, and vieA) of a hypervirulent V. cholerae clinical isolate, MQ1795. Herein, all SNPs and SNP combinations of interest were introduced into the prototypical El Tor reference strain N16961, and the effects on the production of numerous virulence-related factors, including cholera toxin (CT), the toxin-coregulated pilus (TCP), and ToxT, were analyzed. Our data show that triple-SNP (hapR hns luxO and hns luxO vieA) and quadruple-SNP combinations produced the greatest increases in CT, TCP, and ToxT production. The hns and hns luxO SNP combinations were sufficient for increased TCP and ToxT production. Notably, the hns luxO vieA triple-SNP combination strain produced TCP and ToxT levels similar to those of MQ1795. Certain SNP combinations (hapR and hapR vieA) had the opposite effect on CT, TCP, and ToxT expression. Interestingly, the hns vieA double-SNP combination strain increased TCP production while decreasing CT production. Our findings suggest that SNPs identified in the four regulatory genes, in various combinations, are associated with increased virulence capabilities observed in V. cholerae clinical isolates. These studies provide insight into the evolution of highly virulent strains. IMPORTANCE Cholera, an infectious disease of the small intestine caused by the aquatic bacterium Vibrio cholerae, often results in vomiting and acute watery diarrhea. If left untreated or if the response is too slow, the symptoms can quickly lead to extreme dehydration and ultimately death of the patient. Recent anecdotal evidence of cholera patients suffering from increasingly severe symptoms and of disease progression at a much higher rate than previously observed has emerged. As recent cholera outbreaks caused by increasingly virulent strains have resulted in higher mortality rates, the need to investigate the mechanism(s) allowing this observed increased virulence is apparent. The significance of our research is in identifying the mechanism for increased virulence capabilities, which will allow the development of a model that will greatly enhance our understanding of cholera disease and V. cholerae pathogenesis, leading to broader biomedical impacts, as cholera serves as a model for other enteric diarrheal diseases.
Collapse
|
46
|
Countering drug resistance, infectious diseases, and sepsis using metal and metal oxides nanoparticles: Current status. Colloids Surf B Biointerfaces 2016; 146:70-83. [PMID: 27259161 DOI: 10.1016/j.colsurfb.2016.05.046] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Revised: 05/12/2016] [Accepted: 05/16/2016] [Indexed: 12/12/2022]
Abstract
One fourth of the global mortalities is still caused by microbial infections largely due to the development of resistance against conventional antibiotics among pathogens, the resurgence of old infectious diseases and the emergence of hundreds of new infectious diseases. The lack of funds and resources for the discovery of new antibiotics necessitates the search for economic and effective alternative antimicrobial agents. Metal and metal oxide nanoparticles including silver and zinc oxide exhibit remarkable antimicrobial activities against pathogens and hence are one of the most propitious alternative antimicrobial agents. These engineered nanomaterials are approved by regulatory agencies such as USFDA and Korea's FITI, for use as antimicrobial agents, supplementary antimicrobials, food packaging, skin care products, oral hygiene, and for fortifying devices prone to microbial infections. Nevertheless, detailed studies, on molecular and biochemical mechanisms underlying their antimicrobial activity are missing. To take the full advantage of this emerging technology selective antimicrobial activity of these nanoparticles against pathogens should be studied. Optimization of these nanomaterials through functionalization to increase their efficacy and biocompatibility is also required. Urgent in vivo studies on the toxicity of nanomaterials at realistic doses are also needed before their clinical translation.
Collapse
|
47
|
Abstract
Vibrio cholerae can switch between motile and biofilm lifestyles. The last decades have been marked by a remarkable increase in our knowledge of the structure, regulation, and function of biofilms formed under laboratory conditions. Evidence has grown suggesting that V. cholerae can form biofilm-like aggregates during infection that could play a critical role in pathogenesis and disease transmission. However, the structure and regulation of biofilms formed during infection, as well as their role in intestinal colonization and virulence, remains poorly understood. Here, we review (i) the evidence for biofilm formation during infection, (ii) the coordinate regulation of biofilm and virulence gene expression, and (iii) the host signals that favor V. cholerae transitions between alternative lifestyles during intestinal colonization, and (iv) we discuss a model for the role of V. cholerae biofilms in pathogenicity.
Collapse
|
48
|
Microbial biofilms in seafood: A food-hygiene challenge. Food Microbiol 2015; 49:41-55. [DOI: 10.1016/j.fm.2015.01.009] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 01/16/2015] [Accepted: 01/18/2015] [Indexed: 11/21/2022]
|
49
|
Ayala JC, Wang H, Silva AJ, Benitez JA. Repression by H-NS of genes required for the biosynthesis of the Vibrio cholerae biofilm matrix is modulated by the second messenger cyclic diguanylic acid. Mol Microbiol 2015; 97:630-45. [PMID: 25982817 DOI: 10.1111/mmi.13058] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Expression of Vibrio cholerae genes required for the biosynthesis of exopolysacchide (vps) and protein (rbm) components of the biofilm matrix is enhanced by cyclic diguanylate (c-di-GMP). In a previous study, we reported that the histone-like nucleoid structuring (H-NS) protein represses the transcription of vpsA, vpsL and vpsT. Here we demonstrate that the regulator VpsT can disrupt repressive H-NS nucleoprotein complexes at the vpsA and vpsL promoters in the presence of c-di-GMP, while H-NS could disrupt the VpsT-promoter complexes in the absence of c-di-GMP. Chromatin immunoprecipitation-Seq showed a remarkable trend for H-NS to cluster at loci involved in biofilm development such as the rbmABCDEF genes. We show that the antagonistic relationship between VpsT and H-NS regulates the expression of the rbmABCDEF cluster. Epistasis analysis demonstrated that VpsT functions as an antirepressor at the rbmA/F, vpsU and vpsA/L promoters. Deletion of vpsT increased H-NS occupancy at these promoters while increasing the c-di-GMP pool had the opposite effect and included the vpsT promoter. The negative effect of c-di-GMP on H-NS occupancy at the vpsT promoter required the regulator VpsR. These results demonstrate that c-di-GMP activates the transcription of genes required for the biosynthesis of the biofilm matrix by triggering a coordinated VpsR- and VpsT-dependent H-NS antirepression cascade.
Collapse
Affiliation(s)
- Julio C Ayala
- University of Alabama at Birmingham, Department of Microbiology, Birmingham, Alabama.,Morehouse School of Medicine, Department of Microbiology, Biochemistry and Immunology, Atlanta, Georgia
| | - Hongxia Wang
- Morehouse School of Medicine, Department of Microbiology, Biochemistry and Immunology, Atlanta, Georgia.,State Key Laboratory for Infectious Disease Prevention and Control and National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Changping, Beijing, China
| | - Anisia J Silva
- Morehouse School of Medicine, Department of Microbiology, Biochemistry and Immunology, Atlanta, Georgia
| | - Jorge A Benitez
- Morehouse School of Medicine, Department of Microbiology, Biochemistry and Immunology, Atlanta, Georgia
| |
Collapse
|
50
|
Abstract
To cause the diarrheal disease cholera, Vibrio cholerae must effectively colonize the small intestine. In order to do so, the bacterium needs to successfully travel through the stomach and withstand the presence of agents such as bile and antimicrobial peptides in the intestinal lumen and mucus. The bacterial cells penetrate the viscous mucus layer covering the epithelium and attach and proliferate on its surface. In this review, we discuss recent developments and known aspects of the early stages of V. cholerae intestinal colonization and highlight areas that remain to be fully understood. We propose mechanisms and postulate a model that covers some of the steps that are required in order for the bacterium to efficiently colonize the human host. A deeper understanding of the colonization dynamics of V. cholerae and other intestinal pathogens will provide us with a variety of novel targets and strategies to avoid the diseases caused by these organisms.
Collapse
Affiliation(s)
- Salvador Almagro-Moreno
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
- * E-mail:
| | - Kali Pruss
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Ronald K. Taylor
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| |
Collapse
|